WO2010078449A2 - Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders - Google Patents

Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders Download PDF

Info

Publication number
WO2010078449A2
WO2010078449A2 PCT/US2009/069852 US2009069852W WO2010078449A2 WO 2010078449 A2 WO2010078449 A2 WO 2010078449A2 US 2009069852 W US2009069852 W US 2009069852W WO 2010078449 A2 WO2010078449 A2 WO 2010078449A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
nhe
fluid
gastrointestinal tract
pharmaceutical composition
Prior art date
Application number
PCT/US2009/069852
Other languages
French (fr)
Other versions
WO2010078449A3 (en
Inventor
Dominique Charmot
Jeffrey W. Jacobs
Michael Robert Leadbetter
Marc Navre
Chris Carreras
Noah Bell
Original Assignee
Ardelyx, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to EP21178147.1A priority Critical patent/EP3939964A1/en
Priority to DK09796924.0T priority patent/DK2384318T3/en
Priority to JP2011543730A priority patent/JP5502106B2/en
Application filed by Ardelyx, Inc. filed Critical Ardelyx, Inc.
Priority to EP17200784.1A priority patent/EP3351248B1/en
Priority to ES09796924.0T priority patent/ES2657938T3/en
Priority to KR1020177021507A priority patent/KR20170091783A/en
Priority to MX2011007024A priority patent/MX2011007024A/en
Priority to BRPI0923861A priority patent/BRPI0923861B8/en
Priority to MX2015004407A priority patent/MX345283B/en
Priority to KR1020227009722A priority patent/KR20220042487A/en
Priority to LTEP09796924.0T priority patent/LT2384318T/en
Priority to KR1020167033454A priority patent/KR101766619B1/en
Priority to KR1020117017941A priority patent/KR101683318B1/en
Priority to AU2009334511A priority patent/AU2009334511C1/en
Priority to CN2009801576148A priority patent/CN102333759A/en
Priority to CA2748607A priority patent/CA2748607A1/en
Priority to SI200931806T priority patent/SI2384318T1/en
Priority to PL09796924T priority patent/PL2384318T3/en
Priority to EP09796924.0A priority patent/EP2384318B1/en
Priority to KR1020207024357A priority patent/KR20200111230A/en
Priority to NO09796924A priority patent/NO2384318T3/no
Publication of WO2010078449A2 publication Critical patent/WO2010078449A2/en
Publication of WO2010078449A3 publication Critical patent/WO2010078449A3/en
Priority to US13/172,394 priority patent/US8541448B2/en
Priority to IL213852A priority patent/IL213852A/en
Priority to US13/826,186 priority patent/US9006281B2/en
Priority to US13/804,752 priority patent/US8969377B2/en
Priority to US14/592,200 priority patent/US9408840B2/en
Priority to US15/402,211 priority patent/US10543207B2/en
Priority to IL250641A priority patent/IL250641B/en
Priority to US16/476,838 priority patent/US20190374533A1/en
Priority to CY20181100147T priority patent/CY1120451T1/en
Priority to HRP20180289TT priority patent/HRP20180289T1/en
Priority to IL259851A priority patent/IL259851B/en
Priority to US16/773,723 priority patent/US11318129B2/en
Priority to US17/711,863 priority patent/US20230031776A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/18Sulfonamides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/662Phosphorus acids or esters thereof having P—C bonds, e.g. foscarnet, trichlorfon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/55Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/10Antioedematous agents; Diuretics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/22Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound oxygen atoms
    • C07C311/29Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound oxygen atoms having the sulfur atom of at least one of the sulfonamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/12Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D215/14Radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/02Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with only hydrogen atoms or radicals containing only carbon and hydrogen atoms, directly attached to carbon atoms of the nitrogen-containing ring; Alkylene-bis-isoquinolines
    • C07D217/04Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with only hydrogen atoms or radicals containing only carbon and hydrogen atoms, directly attached to carbon atoms of the nitrogen-containing ring; Alkylene-bis-isoquinolines with hydrocarbon or substituted hydrocarbon radicals attached to the ring nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/12Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with radicals, substituted by hetero atoms, attached to carbon atoms of the nitrogen-containing ring
    • C07D217/14Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with radicals, substituted by hetero atoms, attached to carbon atoms of the nitrogen-containing ring other than aralkyl radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/12Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with radicals, substituted by hetero atoms, attached to carbon atoms of the nitrogen-containing ring
    • C07D217/14Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with radicals, substituted by hetero atoms, attached to carbon atoms of the nitrogen-containing ring other than aralkyl radicals
    • C07D217/16Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with radicals, substituted by hetero atoms, attached to carbon atoms of the nitrogen-containing ring other than aralkyl radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/28Phosphorus compounds with one or more P—C bonds
    • C07F9/38Phosphonic acids RP(=O)(OH)2; Thiophosphonic acids, i.e. RP(=X)(XH)2 (X = S, Se)
    • C07F9/3804Phosphonic acids RP(=O)(OH)2; Thiophosphonic acids, i.e. RP(=X)(XH)2 (X = S, Se) not used, see subgroups
    • C07F9/3808Acyclic saturated acids which can have further substituents on alkyl
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/28Phosphorus compounds with one or more P—C bonds
    • C07F9/38Phosphonic acids RP(=O)(OH)2; Thiophosphonic acids, i.e. RP(=X)(XH)2 (X = S, Se)
    • C07F9/3804Phosphonic acids RP(=O)(OH)2; Thiophosphonic acids, i.e. RP(=X)(XH)2 (X = S, Se) not used, see subgroups
    • C07F9/3834Aromatic acids (P-C aromatic linkage)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/28Phosphorus compounds with one or more P—C bonds
    • C07F9/38Phosphonic acids RP(=O)(OH)2; Thiophosphonic acids, i.e. RP(=X)(XH)2 (X = S, Se)
    • C07F9/3804Phosphonic acids RP(=O)(OH)2; Thiophosphonic acids, i.e. RP(=X)(XH)2 (X = S, Se) not used, see subgroups
    • C07F9/3882Arylalkanephosphonic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/576Six-membered rings
    • C07F9/5765Six-membered rings condensed with carbocyclic rings or carbocyclic ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/576Six-membered rings
    • C07F9/60Quinoline or hydrogenated quinoline ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65583Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system each of the hetero rings containing nitrogen as ring hetero atom
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present disclosure is directed to compounds that are substantially active in the gastrointestinal tract to inhibit NHE-mediated antiport of sodium ions and hydrogen ions, and the use of such compounds in the treatment of disorders associated with fluid retention or salt overload and in the treatment of gastrointestinal tract disorders, including the treatment or reduction of pain associated with a gastrointestinal tract disorder
  • CHF congestive heart failure
  • neurohumoral compensatory mechanisms i e , the sympathetic nervous system and the renin-angiotensin system
  • the remn-angiotensm system is activated in response to decreased cardiac output, causing increased levels of plasma renin, angiotensin II, and aldosterone
  • cardiac output increases proportionally, to a point where the heart is unable to dilate further
  • contractility is reduced, so the heart operates at higher volumes and higher filling pressures to maintain output
  • Filling pressures may eventually increase to a level that causes transudation of fluid into the lungs and congestive symptoms (e g , edema, shortness of breath) All of these symptoms are related to fluid volume and salt retention, and this chronic fluid and salt overload further contnbute to disease progression
  • Compliance with the medication regimen and with dietary sodium rest ⁇ ctions is a c ⁇ tical component of self-management for patients with heart failure and may lengthen life, reduce hospitalizations and improve quality of life Physicians often recommend keeping salt intake below 2 3 g per day and no more than 2 g per day for people with heart failure Most people eat considerably more than this, so it is likely that a person with congestive heart failure will need to find ways to reduce dietary salt
  • CHF cardiac output
  • thiazides Thiazides inhibit NaCl transport in the kidney, thereby preventing reabsorption of Na in the cortical diluting segment at the ending portion of the loop of Henle and the proximal portion of the distal convoluted tubule
  • GFR glomerular filtration rate
  • thiazides, as well as other diuretics may cause hypokalemia
  • loop diuretics e g , furosemide
  • Angiotensm-converting enzyme (“ACE”) inhibitors are an example of another drug therapy that may be used to treat congestive heart failure ACE inhibitors cause vasodilatation by blocking the renin-angiotensin-aldosterone system Abnormally low cardiac output may cause the renal system to respond by releasing renin, which then converts angiotensinogen into angiotensin I ACE converts angiotensin I into angiotensin II Angiotensin II stimulates the thirst centers in the hypothalamus and causes vasoconst ⁇ ction, thus increasing blood pressure and venous return Angiotensin II also causes aldosterone to be released, causing reabsorption of Na and concomitant passive reabsorption of fluid, which in turn causes the blood volume to increase ACE inhibitors block this compensatory system and improve cardiac performance by decreasing systemic and pulmonary vascular resistance ACE inhibitors have shown survival benefit and conventionally have been a treatment of choice for CHF However, since ACE inhibitors lower aldo
  • ESRD end stage renal disease
  • i e stage 5 chronic kidney failure
  • the quasi-absence of renal function and ability to eliminate salt and fluid results m large fluctuations in body weight as fluid and salt build up in the body (sodium/volume overload)
  • the fluid overload is characte ⁇ zed as mterdialytic weight gain
  • High fluid overload is also worsened by heart dysfunction, specifically CHF Dialysis is used to remove uremic toxins and also adjust salt and fluid homeostasis
  • symptomatic mtradialytic hypotension may occur when patients are over-dialyzed SIH is exhibited in about 15% to 25% of the ESRD population (Davenport, A , C Cox, and R Thuraisingham, Blood pressure control and symptomatic mtradialytic hypotension in diabetic haemodialysis patients a cross-sectional survey, Nephron CIm Pract , v 109, no 2, p c65-c71 (2008))
  • SIH symptomatic mtradialytic hypo
  • salt and fluid accumulation cont ⁇ bute to the morbidity and mortality of many diseases, including heart failure (in particular, congestive heart failure), chronic kidney disease, end-stage renal disease, liver disease and the like It is also accepted that salt and fluid accumulation are ⁇ sk factors for hypertension Accordingly, there is a clear need for a medicament that, when administered to a patient in need, would result in a reduction in sodium retention, fluid retention, or preferably both Such a medicament would more preferably also not involve or otherwise impair renal mechanisms of fluid/Na homeostasis
  • Diarrhea may be triggered by several agents including, for example, laxatives such as sorbitol, polyethyleneglycol, bisacodyl and phenolphthaleme Sorbitol and polyethyleneglycol triggers osmotic diarrhea with low levels of secreted electrolytes, thus, their utility in removing sodium salt from the GI tract is limited
  • laxatives such as sorbitol, polyethyleneglycol, bisacodyl and phenolphthaleme Sorbitol and polyethyleneglycol triggers osmotic diarrhea with low levels of secreted electrolytes, thus, their utility in removing sodium salt from the GI tract is limited
  • the mechanism of action of phenolphthalem is not clearly established, but is thought to be caused by inhibition of the Na/K ATPase and the CI/HCO 3 anion exchanger and stimulation of electrogenic anion secretion (see, e g , Eherer, A J , C A Santa Ana, J Porter, and J S
  • a fluid-absorbmg polymer such as the natural plant fiber psyllium Polymeric mate ⁇ als, and more specifically hydrogel polymers
  • a fluid-absorbmg polymer such as the natural plant fiber psyllium Polymeric mate ⁇ als, and more specifically hydrogel polymers
  • GI gastrointestinal
  • cation exchange resins have very limited use as drugs, due at least in part to their limited capacity and poor cation binding selectivity Additionally, du ⁇ ng the ion-exchange process, the resms may release a stochiomet ⁇ c amount of exogenous cations (e g , H, K, Ca), which may in turn potentially cause acidosis (H), hyperkalemia
  • exogenous cations e g , H, K, Ca
  • Constipation is characterized by infrequent and difficult passage of stool and becomes chrome when a patient suffers specified symptoms for over 12 non- consecutive weeks withm a 12-month period Chrome constipation is idiopathic if it is not caused by other diseases or by use of medications
  • An evidence-based approach to the management of chronic constipation in North Amenca (Brandt et al , 2005, Am J Gastroenterol 100(Su ⁇ l 1) S5-S21) revealed that prevalence is approximately 15% of the general population Constipation is reported more commonly in women, the elderly, non-whites, and individuals from lower socioeconomic groups
  • IBS Irritable bowel syndrome
  • Constipation is commonly found in the ge ⁇ atnc population, particularly patients with osteoporosis who have to take calcium supplements Calcium supplements have shown to be beneficial in ostoporotic patients to restore bone density but compliance is poor because of calcium-induced constipation effects
  • Opioid-induced constipation (also referred to as opioid-mduced bowel dysfunction or opioid bowel dysfuntion (OBD)) is a common adverse effect associated with opioid therapy OIC is commonly descnbed as constipation, however, it is a constellation of adverse gastrointestinal (GI) effects, which also includes abdominal cramping, bloating, and gastroesophageal reflux Patients with cancer may have disease-related constipation, which is usually worsened by opioid therapy However, OIC is not limited to cancer patients A recent survey of patients taking opioid therapy for pam of non cancer origin found that approximately 40% of patients expe ⁇ enced constipation related to opioid therapy ( ⁇ 3 complete bowel movements per week) compared with 7 6% in a control group Of subjects who required laxative therapy, only 46% of opioid-treated patients (control subjects, 84%) reported achieving the desired treatment results >50% of the time (Pappagallo, 2001, Am J Surg 182(5A Suppl ) 11S-18S) Some patients suffe
  • a major function of the GI tract is to maintain water/Na homeostasis by absorbing virtually all water and Na to which the GI tract is exposed
  • the epithelial layer covering the apical surface of the mammalian colon is a typical electrolyte- transporting epithelium, which is able to move large quantities of salt and water in both directions across the mucosa
  • each day the GI tract processes about 9 liters of fluid and about 800 meq of Na See, e g , Zachos et al , Molecular physiology of intestinal Na+/H+ exchange, Annu Rev Physiol , v 67, p 411-443 (2005) )
  • Only about 1 5 liters of this fluid and about 150 meq of this sodium originates from ingestion, rather, the majo ⁇ ty of the fluid (e g , about 7 5 liters) and sodium (about 650 meq) is secreted via the GI organs as part of digestion
  • Plasma membrane NHEs cont ⁇ bute to maintenance of intracellular pH and volume, transcellular absorption of NaCl and NaHCC> 3 , and fluid balance earned out by epithelial cells, especially in the kidney, intestine, gallbladder, and salivary glands, as well as regulation of systemic pH
  • a body of literature devoted to the role and clinical intervention on systemic NHEs to treat disorders related to ischemia and reperfusion for cardioprotection or renal protection
  • NHE 2, NHE 3 and NHE 8 are expressed on the apical side of the GI tract, with NHE 3 providing a larger contribution to transport
  • NHE 2 NHE 3 and NHE 8 are expressed on the apical side of the GI tract
  • the present invention is directed to compounds that are substantially active in the gastrointestinal tract to inhibit NHE-mediated antiport of sodium ions and hydrogen ions, and the use of such compounds in the treatment of disorders associated with fluid retention and salt overload and in the treatment of gastrointestinal tract disorders, including the treatment or reduction of pam associated with a gastrointestinal tract disorder
  • a compound is provided having (l) a topological
  • tPSA Polar Surface Area
  • tPSA tPSA
  • the compound is substantially active in the gastrointestinal tract to inhibit NHE- mediated antiport of sodium ions and hydrogen ions therein upon administration to a patient in need thereof
  • the compound has a molecular weight of at least about 500 Da, at least about 1000 Da, at least about 2500 Da, or at least about about 5000 Da
  • the compound has a tPS A of at least about 250 A 2 , at least about 270 A 2 , at least about 300 A 2 , at least about 350 A 2 , at least about 400 A 2 , or at least about 500 A 2
  • the compound is substantially active on the apical side of the epithelium of the gastrointestinal tract to inhibit antiport of sodium ions and hydrogen ions mediated by NHE-3, NHE-2, NHE-8, or a combination thereof
  • the compound is substantially systemically non-bioavailable and/or substantially impermeable to the epithelium of the gastrointestinal tract
  • the compound is substantially active in the lower gastrointestinal tract
  • the compound has (i) a total number of NH and/or OH and/or other potential hydrogen bond donor moieties greater than about 5, (ii) a total number of O atoms and/or N atoms and/or other potential hydrogen bond acceptors greater than about 10, and/or (in) a Moiiguchi partition coefficient greater than about 10 5 or less than about 10
  • the compound has a permeability coefficient, P app , of less than about 100 x 10 6 cm/s, or less than about 10 x 10 6 cm/s, or less than about
  • the compound has a structure of Formula (I) or (IX)
  • NHE is a NHE-mhibitmg small molecule that comp ⁇ ses (1) a hetero- atom containing moiety, and (11) a cyclic or heterocyclic scaffold or support moiety bound directly or indirectly thereto, the heteroatom-containing moiety being selected from a substituted guamdmyl moiety and a substituted heterocyclic moiety, which may optionally be fused with the scaffold or support moiety to form a fused bicyclic structure, and,
  • Z is a moiety having at least one site thereon for attachment to the NHE- inhibiting small molecule, the resulting NHE-Z molecule possessing overall physicochemical properties that render it substantially impermeable or substantially systemically non bioavailable, and, E is an integer having a value of 1 or more
  • the total number of freely rotatable bonds m the total number of freely rotatable bonds
  • NHE-Z molecule is at least about 10 In further embodiments, the total number hydrogen bond donors in the NHE-Z molecule is at least about 5 In further embodiments, the total number of hydrogen bond acceptors in the NHE-Z molecule is at least about 10 In further embodiments, the total number of hydrogen bond donors and hydrogen bond acceptors m the NHE-Z molecule is at least about 10 In further embodiments, the Log P of the NHE-Z inhibiting compound is at least about 5 In further embodiments, the log P of the NHE-Z inhibiting compound is less than about 1, or less than about 0 In further embodiments, the scaffold is a 5-member or 6-member cyclic or heterocyclic moiety In further embodiments, the scaffold is aromatic
  • the scaffold of the NHE-inhibitmg small molecule is bound to the moiety, Z, and the compound has the structure of Formula (II)
  • Z is a Core having one or more sites thereon for attachment to one or more NHE-mhibitmg small molecules, the resulting NHE-Z molecule possessing overall physicochemical properties that render it substantially impermeable or substantially systemically non-bioavailable
  • B is the heteroatom-containing moiety of the NHE-inhibitmg small molecule, and is selected from a substituted guanidinyl moiety and a substituted heterocyclic moiety, which may optionally be fused with the Scaffold moiety to form a fused, bicyclic structure
  • Scaffold is the cyclic or heterocyclic scaffold or support moiety of the NHE-inhibitmg small molecule, which is bound directly or indirectly to heteroatom- containing moiety, B, and which is optionally substituted with one or more additionally hydrocarbyl or heterohydrocarbyl moieties,
  • X is a bond or a spacer moiety selected from a group consisting of substituted or unsubstituted hydrocarbyl or heterohydrocarbyl moieties, and in particular substituted or unsubstituted Ci 7 hydrocarbyl or heterohydrocarbyl, and substituted or unsubstituted, saturated or unsaturated, cyclic or heterocyclic moieties, which links B and the Scaffold, and,
  • D and E are integers, each independently having a value of 1 or more
  • the compound is an oligomer, dend ⁇ mer or polymer
  • Z is a Core moiety having two or more sites thereon for attachment to multiple NHE-mhibitmg small molecules, either directly or indirectly through a linking moiety, L, and the compound has the structure of Formula (X)
  • L is a bond or linker connecting the Core to the NHE-inhibitmg small molecule
  • n is an integer of 2 or more, and further wherein each NHE- inhibiting small molecule may be the same or differ from the others
  • the NHE-mhibiting small molecule has the structure of Formula (IV)
  • each Ri, R 2 , R 3 , R 5 and R 9 are independently selected from H, halogen, -
  • NR 7 (CO)R 8 -(CO)NR 7 R 8 , -SO 2 -NR 7 R 8 , -NR 7 SO 2 R 8 , -NR 7 R 8 , -OR 7 , -SR 7 , - 0(CO)NR 7 R 8 , -NR 7 (CO)OR 8 , and -NR 7 SO 2 NR 8 , where R 7 and R 8 are independently selected from H or a bond linking the NHE-mhibitmg small molecule to L, provided at least one is a bond linking the NHE-inhibihng small molecule to L, R 4 is selected from H, Ci-C 7 alkyl, or a bond linking the NHE-mhibiting small molecule to L,
  • R. 6 is absent or selected from H and Ci-C 7 alkyl
  • ArI and Ar2 independently represent an aromatic ⁇ ng or a heteroaromatic nng
  • the NHE-mhibitmg small molecule has the following structure
  • each R 1 , R 2 and R 3 are independently selected from H, halogen, -
  • R 7 and R 8 are independently selected from H or a bond linking the NHE-inhibiting small molecule to L, provided at least one is a bond linking the NHE-inhibitmg small molecule to L
  • the NHE-inhibiting small molecule has one of the following structures
  • L is a polyalkylene glycol linker In further embodiments, L is a polyethylene glycol linker In further embodiments, n is 2
  • the Core has the following structure
  • Ci salkylene optionally substituted aryl, optionally substituted heteroaryl, a polyethylene glycol linker, -(CH 2 )i 6 ⁇ (CH 2 )i 6- and -(CH 2 )i 6 NY,(CH 2 )i 6 -, and
  • Yi is selected from the group consisting of hydrogen, optionally substituted Ci salkyl, optionally substituted aryl or optionally substituted heteroaryl
  • the Core is selected from the group consisting
  • the compound is an oligomer
  • Z is a linking moiety, L, that links two or more NHE-mhibitmg small molecules together, when the two or more NHE-inhibiting small molecules may be the same or different, and the compound has the structure of Formula (XI)
  • L is a bond or linker connecting one NHE-mhibiting small molecule to another, and m is 0 or an integer of 1 or more
  • the compound is an oligomer, dendnmer or polymer, and Z is a backbone, denoted Repeat Unit, to which is bound multiple NHE- inhibiting moieties, and the compound has the structure of Formula (XIIB)
  • a pharmaceutical composition comprising a compound as set forth above, or a stereoisomer, pharmaceutically acceptable salt or prodrug thereof, and a pharmaceutically acceptable earner, diluent or excipient
  • the composition further comprises a fluid- absorbing polymer
  • the flmd-absorbmg polymer is delivered directly to the colon
  • the fluid-absorbing polymer has a fluid absorbency of at least about 15 g of isotonic fluid per g of polymer under a static pressure of about 5 kPa
  • the fluid-absorbing polymer has a fluid absorbency of at least about 15 g of isotonic fluid per g of polymer under a static pressure of about 10 kPa
  • the fluid-absorbing polymer is characterized by a fluid absorbency of at least about 10 g/g
  • the fluid-absorbing polymer is characte ⁇ zed by a fluid absorbency of at least about 15 g/g
  • the fluid-absorbing polymer is superabsorbent
  • the fluid absorbing polymer is a crosshnked, partially neutralized polyelectrolyte hydrogel
  • the fluid absorbing polymer is a crosshnked, partially neutralized polyelectrolyte hydrogel
  • the composition further comprises another pharmaceutically active agent or compound
  • the composition further comp ⁇ ses another pharmaceutically active agent or compound selected from the group consisting of a diuretic, cardiac glycoside, ACE inhibitor, angiotensm-2 receptor antagonist, calcium channel blocker, beta blocker, alpha blocker, central alpha agonist, vasodilator, blood thinner, anti-platelet agent, hpid-lowe ⁇ ng agent, and peroxisome prohferator-activated receptor (PPAR) gamma agonist agent
  • the diuretic is selected from the group consisting of a high ceiling loop diuretic, a benzothiadiazide diuretic, a potassium spa ⁇ ng diuretic, and a osmotic diuretic
  • the composition further comp ⁇ ses another pharmaceutically active agent or compound selected from the group consisting of an analgesic peptide or agent
  • the composition further comp ⁇ ses another pharmaceutically active agent or compound selected from the group consisting of a la
  • a method for inhibiting NHE-mediated antiport of sodium and hydrogen ions is provided, the method composing administering to a mammal in need thereof a pharmaceutically effective amount of a compound or pharmaceutical composition as set forth above
  • a method for treating a disorder associated with fluid retention or salt overload is provided, the method composing administering to a mammal in need thereof a pharmaceutically effective amount of a compound or pharmaceutical composition as set forth above
  • a method for treating a disorder selected from the group consisting of heart failure (such as congestive heart failure), chrome kidney disease, end-stage renal disease, liver disease, and peroxisome proliferator-activated receptor (PPAR) gamma agonist-induced fluid retention is provided, the method comp ⁇ sing administering to a mammal in need thereof a pharmaceutically effective amount of a compound or pharmaceutical composition as set forth above
  • a method for treating hypertension comp ⁇ sing administering to a mammal in need thereof a pharmaceutically effective amount of a compound or pharmaceutical composition as set forth above
  • the method comp ⁇ ses administering a pharmaceutically effective amount of the compound to the mammal in order to increase the mammal's daily fecal output of sodium and/or fluid
  • the method comp ⁇ ses administe ⁇ ng a pharmaceutically effective amount of the compound to the mammal in order to increase the mammal's daily fecal output of sodium by at least about 30 mmol, and/or fluid by at least about 200 ml hi further embodiments, the mammal's fecal output of sodium and/or fluid is increased without introducing another type of cation m a stoichiomet ⁇ c or near stoichiomet ⁇ c fashion via an ion exchange process
  • the method further comp ⁇ ses administering to the mammal a fluid-absorbing polymer to absorb fecal fluid resulting from the use of the compound that is substantially active m the gastrointestinal tract to inhibit NHE- mediated antiport of sodium ions and hydrogen ions therein
  • the compound or composition is administered to treat hypertension In further embodiments, the compound or composition is administered to treat hypertension associated with dietary salt intake In further embodiments, administration of the compound or composition allows the mammal to intake a more palatable diet In further embodiments, the compound or composition is administered to treat fluid overload In further embodiments, the fluid overload is associated with congestive heart failure In further embodiments, the fluid overload is associated with end stage renal disease In further embodiments, the fluid overload is associated with peroxisome prohferator-activated receptor (PPAR) gamma agonist therapy In further embodiments, the compound or composition is administered to treat sodium overload In further embodiments, the compound or composition is administered to reduce mterdialytic weight gam m ESRD patients In further embodiments, the compound or composition is administered to treat edema In further embodiments, the edema is caused by chemotherapy, pre-menstrual fluid overload or preeclampsia In further embodiments, the compound or composition is administered orally, by rectal suppository, or
  • the method comp ⁇ ses administering a pharmaceutically effective amount of the compound or composition in combination with one or more additional pharmaceutically active compounds or agents
  • the one or more additional pharmaceutically active compounds or agents is selected from the group consisting of a diuretic, cardiac glycoside, ACE inhibitor, angiotensin-2 receptor antagonist, aldosterone antagonist, calcium channel blocker, beta blocker, alpha blocker, central alpha agonist, vasodilator, blood thinner, anti-platelet agent, hpid-lowe ⁇ ng agent, and peroxisome prohferator-activated receptor (PPAR) gamma agonist agent
  • the diuretic is selected from the group consisting of a high ceiling loop diuretic, a benzothiadiazide diuretic, a potassium spa ⁇ ng diuretic, and a osmotic diuretic
  • the pharmaceutically effective amount of the compound or composition, and the one or more additional pharmaceutically active compounds or agents are administered as part of a single
  • a method for treating irritable bowel syndrome comprising administering to a mammal in need thereof a pharmaceutically effective amount of an NHE-3 inhibitor compound or a pharmaceutical composition composing an NHE-3 inhibitor compound.
  • the NHE-3 inhibitor compound or the pharmaceutical composition composing an NHE-3 inhibitor compound is a compound or pharmaceutical composition as set forth above
  • the compound or composition is administered to treat or reduce pain associated with a gastrointestinal5 tract disorder In further embodiments, the compound or composition is administered to treat or reduce visceral hypersensitivity associated with a gastrointestinal tract disorder In further embodiments, the compound or composition is administered to treat or reduce inflammation of the gastrointestinal tract In further embodiments, the compound or composition is administered to reduce gastrointestinal transit time 0 In further embodiments, the compound or composition is administered either orally or by rectal suppository
  • the method composes admmisteong a pharmaceutically effective amount of the compound or composition, in combination with one or more additional pharmaceutically active compounds or agents
  • the one or more additional pharmaceutically active agents or compounds are an analgesic peptide or agent
  • the one or more additional pharmaceutically active agents or compounds are selected from the group consisting of a laxative agent selected from a bulk-producing agent (e g psyllium husk (Metamucil)), methylcellulose (Citrucel), polycarbophil, dietary fiber, apples, stool0 softeners/surfactant (e g , docusate, Colace, Diocto), a hydrating or osmotic agent (e g , dibasic sodium phosphate, magnesium citrate, magnesium hydroxide (Milk of magnesia), magnesium sulfate (which is Epsom salt), monobasic sodium phosphate, sodium biphosphate), and a hyperosmotic agent (e
  • a laxative agent selected from a
  • Figure 1 is a graph that illustrates the relationship between tPSA and
  • FIGs 2A and 2B are graphs that illustrate the cecum and colon water content after oral administration of certain example compounds, as further discussed in the Examples (under the subheading "3 Pharmacological Test Example 3")
  • Figures 3A and 3B are graphs that illustrate the dose dependent decrease of u ⁇ nary salt levels after administration of certain example compounds, as further discussed in the Examples (under the subheading "14 Pharmacological Test Example
  • Figure 4 is a graph that illustrates a dose dependent increase in fecal water content after administration of a certain example compound, as further discussed in the Examples (under the subheading "15 Pharmacological Test Example 15")
  • Figures 5A, 5B and 5C are graphs that illustrate that supplementing the diet with Psyllium results in a slight reduction of fecal stool form, but without impacting the ability of a certain example compound to increase fecal water content or decrease u ⁇ nary sodium, as further discussed in the Examples (under the subheading "16 Pharmacological Test Example 16")
  • Figure 6 is a graph that illustrates that inhibition of NHE-3 reduces hypersensitivity to distention, as further discussed in the Examples (under the subheading "17 Pharmacological Test Example 17")
  • Figures 7A and 7B are graphs that illustrate that inhibition of NHE-3 increases the amount of sodium excreted in feces, as further discussed in the Examples (under subheading "18 Pharmacological Test Example 18")
  • NHE-mediated antiport of sodium ions (Na + ) and hydrogen ions (H + ) in the gastrointestinal tract is a powerful approach to the treatment of va ⁇ ous disorders that may be associated with or caused by fluid retention and/or salt overload, and/or disorders such as heart failure (in particular, congestive heart failure), chrome kidney disease, end-stage renal disease, liver disease, and/or peroxisome prohferator-activated receptor (PPAR) gamma agomst-mduced fluid retention
  • heart failure in particular, congestive heart failure
  • PPAR peroxisome prohferator-activated receptor
  • NHE-mediated antiport of sodium ions (Na + ) and hydrogen ions (H + ) in the gastrointestinal tract, and more particularly the gastrointestinal epithelia is a powerful approach to the treatment of hypertension, that may be associated with or caused by fluid retention and/or salt overload More specifically, it has been found that the inhibition of the NHE-mediated antiport of sodium ions and hydrogen ions in the GI tract increases the fecal excretion of sodium, effectively reducing systemic levels of sodium and fluid This, in turn, improves the clinical status of a patient suffe ⁇ ng from hypertension
  • Such a treatment may optionally be enhanced by the co-admimstration of other beneficial compounds or compositions, such as for example a fluid-absorbing polymer
  • the fluid-absorbmg polymer may optimally be chosen so that it does not block or otherwise negatively interfere with the mechanism of action of the co-dosed NHE inhibitor and/or hypertension
  • NHE-mediated antiport of sodium ions (Na + ) and hydrogen ions (H + ) in the gastrointestinal tract, and more particularly the gastrointestinal epithelia is a powerful approach to the treatment of va ⁇ ous gastrointestinal tract disorders, including the treatment or reduction of pam associated with gastrointestinal tract disorders, and more particularly to the restoration of appropnate fluid secretion in the gut and the improvement of pathological conditions encountered m constipation states
  • Applicants have further recognized that by blocking sodium ion re-absorption, the compound of the invention restore fluid homeostasis in the GI tract, particularly in situations wherein fluid secretion/absorption is altered in such a way that it results in a high degree of feces dehydration, low gut motility, and/or a slow transit-time producing constipation states and GI discomfort generally
  • such a treatment may optionally be enhanced by the co- administration of other beneficial compounds or compositions, such as for
  • the method of the present disclosure employs the use of compounds and compositions that are desirably highly selective or localized, thus acting substantially in the gastrointestinal tract without exposure to other tissues or organs In this way, any systemic effects can be minimized (whether they are on-target or off-target) Accordingly, it is to be noted that, as used herein, and as further detailed elsewhere herein, “substantially active in the gastrointestinal tract” generally refers to compounds 5 that are substantially systemically non-bioavailable and/or substantially impermeable to the layer of epithelial cells, and more specifically epithelium of the GI tract It is to be further noted that, as used herein, and as further detailed elsewhere herein, “substantially impermeable” more particularly encompasses compounds that are impermeable to the layer of epithelial cells, and more specifically the gastrointestinal0 epithelium (or epithelial layer) "Gastrointestinal epithelium” refers to the membranous tissue
  • substantially systemically non-bioavailable generally refers to the inability to detect a compound in the systemic circulation of an animal or human following an oral dose of the compound
  • a compound to be bioavailable it must be transferred across the gastrointestinal epithelium (that is, substantially permeable as defined above), be transported via the portal circulation to the liver, avoid substantial metabolism in the liver, and then be transferred into systemic circulation
  • small molecules exhibiting an inhibitory effect on NHE-mediated antiport of sodium and hydrogen ions desc ⁇ bed herein may be modified or functionahzed to render them "substantially active” in the GI tract (or “substantially impermeable” to the GI tract and/or “substantially systemically non-bioavailable” from the GI tract) by, for example, ensunng that the final compound has (i) a molecular weight of greater than about 500 Daltons (Da) (e g , greater than about 1000 Da,
  • One or more of the above-noted methods for structurally modifying or functionahzing the NHE-mhibiting small molecule may be utilized in order to prepare a compound suitable for use in the methods of the present disclosure, so as to render the compound substantially impermeable or substantially systemically non-bioavailable, that is, one or more of the noted exemplary physical properties may be "engineered" into the NHE-mhibiting small molecule to render the resulting compound substantially impermeable or substantially systemically non-bioavailable, or more generally substantially active, in the GI tract, while still possessing a region or moiety therein that is active to inhibit NHE-mediated antiport of sodium ions and hydrogen ions
  • the NHE-inhibitors e g , NHE-3, -2 and/or -8) of the instant disclosure are believed to act via a distinct and unique mechanism, causing the retention of fluid and ions in the GI tract (and stimulating fecal excretion) rather than stimulating increased secretion of said fluid and ions
  • lubiprostone Amitiza® Sucampo/Takeda
  • ClC-2 Type 2 Chlo ⁇ de Channel
  • Lmaclotide MD-1100 acetate, Microbia/Forest Labs
  • ClC-2 Type 2 Chlo ⁇ de Channel
  • the present disclosure encompasses essentially any small molecule, which may be monovalent or polyvalent, that is effective or active as a
  • NHE inhibitor and that is substantially active in the GI tract, and more particularly substantially impermeable or substantially systemically non-bioavalable therein, including known NHE inhibitors that may be modified or functionalized in accordance with the present disclosure to alter the physicochemical properties thereof so as to render the overall compound substantially active in the GI tract
  • present disclosure encompasses monovalent or polyvalent compounds that are effective or active as NHE-3 , NHE-2 and/or NHE-8 inhibitors
  • NHE represents a NHE-inhibitmg small molecule
  • Z represents a moiety having at least one site thereon for attachment to an NHE-mhibitmg small molecule, the resulting NHE-Z molecule possessing overall physicochemical properties that render it substantially impermeable or substantially systemically non-bioavailable
  • the NHE-inhibitmg small molecule generally comp ⁇ ses a heteroatom-contaimng moiety and a cyclic or heterocyclic scaffold or support moiety bound directly or indirectly thereto
  • examination of the structures of small molecules reported to-date to be NHE inhibitors suggest, as further illustrated herein below, that most comp ⁇ se a cyclic or heterocyclic support or scaffold bound directly or indirectly (by, for example, an acyl moiety or a hydrocabyl or heterohydrocarbyl moiety, such as an alkyl, an alkenyl, a heteroalkyl or a heteroalkenyl moiety) to a
  • the Z moiety may be bound to essentially any position on, or within, the NHE small molecule, and in particular may be (i) bound to the scaffold or support moiety, (ii) bound to a position on, or withm, the heteroatom-contaimng moiety, and/or (m) bound to a position on, or within, a spacer moiety that links the scaffold to the5 heteroatom-contaimng moiety, provided that the installation of the Z moiety does not significantly adversely impact NHE-inhibitmg activity
  • Z may be in the form of an oligomer, dend ⁇ mer or polymer bound to the NHE small molecule (e g , bound for example to the scaffold or the spacer moiety), or alternatively Z may be in the form of a linker that links multiple NHE small molecules together, and0 therefore that acts to increase (i) the overall molecular weight and/or polar surface area of the NHE-Z molecule, and/or, (ii) the number of freely rotatable
  • the present disclosure is more particularly directed to such a substantially impermeable or substantially systemically non-bioavailable, NHE- mhibiting compound, or a pharmaceutical salt thereof, wherein the compound has the0 structure of Formula (II) Substantially Impermeable and/or substantially systemically non-bioavailable
  • (i) Z is a moiety bound to or incorporated in the NHE-inhibiting small molecule, such that the resulting NHE-Z molecule possesses overall physicochemical properties that render it substantially impermeable or substantially systemically non-bioavailable
  • B is the heteroatom-contaimng moiety of the NHE-inhibiting small molecule
  • m one particular embodiment is selected from a substituted guamdinyl moiety and a substituted heterocyclic moiety, which may optionally be fused with the Scaffold moiety to form a fused, bicychc structure
  • (m) Scaffold is the cyclic or heterocyclic moiety to which is bound directly or indirectly the hetero-atom containing moiety (e g , the substituted guamdinyl moiety or a substituted heterocyclic moiety), B, and which is optionally substituted with one or more additionally hydrocarbyl or heterohydrocarbyl moieties
  • X is the cyclic or heterocyclic moiety to which
  • B may be selected from a guamdinyl moiety or a moiety that is a guamdinyl bioisostere selected from the group consisting of substituted cyclobutenedione, substituted imidazole, substituted thiazole, substituted oxadiazole, substituted pyrazole, or a substituted amine More particularly, B may be selected from guamdinyl, acylguanidinyl, sulfonylguamdinyl, or a guamdine bioisostere such as a cyclobutenedione, a substituted or unsubstituted 5- or 6-member heterocycle such as substituted or unsubstituted imidazole, aminoimidazole, alkyhmidizole, thiazole, oxadiazole, pyrazole, alkylthioimidazole, or other functionality that may optionally become positively charged or function as a sodium
  • bioisostere generally refers to a moiety with similar physical and chemical properties to a guamdme moiety, which in turn imparts biological properties to that given moiety similar to, again, a guamdme moiety, in this instance (See, for example, Ahmad, S et al , Ammoimidazoles as Bioisosteres of Acylguanidines Novel, Potent, Selective and Orally Bioavailable Inhibitors of the Sodium Hydrogen Exchanger Isoform-1, Boorganic & Med Chem Lett , pp 177-180 (2004), the entire contents of which is incorporated herein by reference for all relevant and consistent purposes )
  • known NHE-mhibiting small molecules or chemotypes that may serve as suitable starting mate ⁇ als (for modification or functionahzation, in order to render the small molecules substantially impermeable or substantially systemically non-bioavailable, and/or used in pharmaceutical preparations in combination with, for example, a fluid-absorbing polymer) may generally be organized into a number of subsets, such as for example
  • terminal ⁇ ng (or, m the case of the non-acyl guamdmes, "R"), represent the scaffold or support moiety
  • the guamdme moiety (or the substituted heterocycle, and more specifically the pipe ⁇ dine ⁇ ng, in the case of the non-guamdine inhibitors) represents B
  • X is the acyl moiety, or the -A-B-acyl- moiety (or a bond in the case of the non-acyl guanidmes and the non-guamdine inhibitors)
  • the heteroatom-containing moiety may be capable of forming a positive charge, this should not be understood or interpreted to require that the overall compound have a net positive charge, or only a single positively charged moiety therein, or even that the heteroatom-contaimng moiety therein be capable of forming a positive charge in all instances Rather, in various alternative embodiments, the compound may have no charged moieties therein, or it may have multiple charged 5 moieties therein (which may have positive charges, negative charges, or a combination thereof) Additionally, it is to be understood that the overall compound may have a net neutral charge, a net positive charge, or a net negative charge
  • bioisostenc replacements for guamdine or acylguanidine may also be used.
  • Potentially viable bioisostenc "guamdine replacements" identified to-date have a five- or six-membered heterocyclic ⁇ ng with donor/acceptor and pKa patterns similar to
  • NHE-mhibitmg compounds for substantially impermeable or substantially systemically non-bioavailable NHE- mhibitmg compounds, and/or for NHE-mhibitmg small molecules suitable for modification or functionalization in accordance with the present disclosure so as to render them substantially impermeable or substantially systemically non-bioavailable
  • various identifiers e g , atom identifiers in a chain or nng, identifiers for substituents on a ⁇ ng or chain, etc
  • An identifier in one structure should therefore not be assumed to have the same meaning in a different structure, unless specifically stated (e g , "Ri" in one structure may or may not be the same as “Ri” in another structure)
  • specific details of the structures, including one or more of the identifiers therein may be provided m a cited reference, the
  • the substantially impermeable or substantially systemically non- bioavailable NHE-inhibiting compounds of the present disclosure may in general be de ⁇ ved or prepared from essentially any small molecule possessing the ability to inhibit NHE activity, including small molecules that have already been reported or identified as inhibiting NHE activity but lack impermeability (i e , are not substantially impermeable)
  • the compounds utilized in the various methods of the present disclosure are de ⁇ ved or prepared from small molecules that inhibit the NHE-3, -2, and/or -8 isoforms To-date, a considerable amount of work has been devoted to the study of small molecules exhibiting NHE-I inhibition, while less has been devoted for example to the study of small molecules exhibiting NHE-3 inhibition
  • the present disclosure is directed generally to substantially impermeable or substantially systemically non-bioavailable NHE-inhibiting compounds, the substantially impermeable or substantially systemically non- bioavailable compounds exhibiting NHE-3, -2, and/or -8 inhibition
  • R. 6 and R 7 are a halogen (e g , Cl), Rs is lower alkyl (e g , CH 3 ), and R 1 -R 4 are H, the compound having for example the structure
  • the following small molecule disclosed in Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and in particular pages 1-2 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification m accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
  • va ⁇ ables in the structure are defined in the cited patent application, the details of which are incorporated herein by reference
  • the following small molecule disclosed in Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and in particular page 49 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification m accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non bioavailable)
  • the following small molecule disclosed m Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and m particular pages 127-129 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
  • the following small molecule disclosed in Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and in particular pages 134-137 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification m accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
  • the following small molecule disclosed in Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and in particular pages 37- 45 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
  • the following small molecule disclosed in Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and in particular pages 90- 91 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
  • the following small molecule disclosed in U S Patent No 5,900,436 (or EP 0822182 Bl), the entire contents of which (and in particular column 1, lines 10-55 therein) are incorporated herein by reference for all relevant and consistent purposes, may be suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or 5 substantially systemically non-bioavailable)
  • va ⁇ ables in the structures are defined in the cited patents, the details of which are C incorporated herein by reference
  • the following small molecule disclosed in Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and in particular pages 35- 47 therein) is incorporated herein for all relevant and consistent purposes, may be5 suitable for use or modification m accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
  • va ⁇ ables in the structure are defined in the cited patent application, the details of which are incorporated herein by reference
  • va ⁇ ables in the structure are defined m the cited patent application, the details of which are incorporated herein by reference
  • the following small molecule,0 disclosed in U S Patent Nos 6,911,453 and 6,703,405, the entire contents of which (and in particular the text of columns 1-7 and 46 of 6,911,453 and columns 14-15 of 6,703,405) are incorporated herein by reference for all relevant and consistent purposes, may be suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is5 substantially impermeable or substantially systemically non-bioavailable)
  • va ⁇ ables in the structure are defined in the cited patents, the details of which areQ incorporated herein by reference
  • a particularly preferred small molecule falling within the above-noted structure is further illustrated below (see, e g , Example 1 of the 6,911,453 patent, the entire contents of which are specifically incorporated herein by reference)
  • the following small molecules disclosed in U S Patent Publication Nos 2004/0039001, 2004/0224965, 2005/0113396 and 2005/0020612, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes, may be suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
  • va ⁇ ables in the structures are defined above and/or m one or more of the cited patent applications, the details of which are incorporated herein by reference, and/or as illustrated above (wherein the broken bonds indicate a point of attachment for the Y moiety to the fused heterocyclic ⁇ ng)
  • the combination of X and Y may be as follows
  • the small molecule has the general structure
  • Ri, R 2 and R 3 may be the same or different, but are preferably different, and are independently selected from H, NR'R" (wherein R' and R" are independently selected from H and hydrocarbyl, such as lower alkyl, as defined elsewhere herein) and the structure )
  • the following small molecule disclosed in U S Patent No 6,399,824, the entire content of which (and in particular the text of Example 1 therein) is incorporated herein by reference for all relevant and consistent purposes, may be particularly suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
  • R may be preferably selected from H and (CHa) 2 NCH 2 CH 2 -, with H being particularly preferred in various embodiments
  • the following small molecule disclosed in U S Patent No 6,005,010 (and in particular columns 1-3 therein), and/or U S Patent No 6,166,002 (and in particular columns 1-3 therein), the entire contents of which are incorporated herein by reference for all relevant and consistent purposes, may be suitable for use or modification m accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
  • the following small molecule disclosed in U S Patent Application No 2008/0194621, the entire content of which (and in particular the text of Example 1 therein) is incorporated herein by reference for all relevant and consistent purposes, may be particularly suitable for use or modification m accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
  • the following small molecule disclosed in U S Patent No 6,911,453, the entire content of which (and m particular the text of Example 35 therein) is incorporated herein by reference for all relevant and consistent purposes, may be particularly suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
  • the small molecule may be selected from the group consisting of
  • a bond or link may extend, for example, between the Core and amme-substituted aromatic ⁇ ng (first structure), the heterocyclic ⁇ ng or the aromatic ⁇ ng to which it is bound, or alternatively the chloro-substituted aromatic nng (second structure), or the difluoro-substituted aromatic ⁇ ng or the sulfonamide- substituted aromatic ⁇ ng (third structure) C.
  • first structure the Core and amme-substituted aromatic ⁇ ng
  • the heterocyclic ⁇ ng or the aromatic ⁇ ng to which it is bound or alternatively the chloro-substituted aromatic nng (second structure), or the difluoro-substituted aromatic ⁇ ng or the sulfonamide- substituted aromatic ⁇ ng (third structure) C.
  • NHE-I inhibitors Shown below are examples of various NHB inhibiting small molecules and their selectivity across the NHE-I, -2 and -3 isoforms (See, e g , B Masereel et al , An Overview of Inhibitors of Na+ / H+ Exchanger, European J of Med Chem , 38, pp 547-554 (2003), the entire contents of which is incorporated by reference here for all relevant and consistent purposes)
  • Most of these small molecules were optimized as NHE-I inhibitors, and this is reflected in their selectivity with respect thereto (IC50's for subtype- 1 are significantly more potent (numerically lower) than for subtype-3)
  • IC50's for subtype- 1 are significantly more potent (numerically lower) than for subtype-3)
  • the data in Table 1 indicates that NHE-3 activity may be engineered into an inhibitor se ⁇ es originally optimized against a different isoform
  • amilo ⁇ de is a poor NHE-3 inhibitor and was inactive against this anti
  • the NHE inhibitor small molecules disclosed herein may advantageously be modified to render them substantially impermeable or substantially systemically non-bioavailable
  • the compounds as descnbed herein are, accordingly, effectively localized in the gastrointestinal tract or lumen, and in one particular embodiment the colon Since the various NHE isomforms may be found in many different internal organs (e g , bram, heart, liver, etc ), localization of the NHE inhibitors in the intestinal lumen is desirable in order to minimize or eliminate systemic effects (i e , prevent or significantly limit exposure of such organs to these compounds)
  • the present disclosure provides NHE inhibitors, and in particular NHE-3, -2 and/or -8 inhibitors, that are substantially systemically non-bioavailable in the GI tract, and more specifically substantially systemically impermeable to the gut epithelium, as further descnbed below
  • the "NHE-Z" molecule is monovalent, that is, the molecule contains one moiety that effectively acts to inhibit NHE-mediated antiport of sodium ions and hydrogen ions
  • the NHE-Z molecule may be selected, for example, from one of the following structures of Formulas (IV), (V), (VI) or (VII)
  • each Ri , R 2 , R 3 , R 5 and R 9 are independently selected from H, halogen (e g , Cl), -NR 7 (CO)R 8 , -(CO)NR 7 R 8 , -SO 2 -NR 7 R 8 , -NR 7 SO 2 R 8 , -NR 7 R 8 , -OR 7 , -SR 7 , - 0(CO)NR 7 R 8 , -NR 7 (CO)OR 8 , and -NR 7 SO 2 NR 8 , where R 7 and R 8 are independently selected from H or Z, where Z is selected from substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, polyalkylene glycol and polyols, where substituents thereon are selected from hydroxyls, amines, amidines, carboxylates, phosphonates, sulfonates, and guamdines, R 4 is selected from H, CrC 7 alkyl or Z, where Z is selected from substitute
  • each Ri, R 2 , R 3 , and R 5 are independently selected from H, -NR 7 (CO)Rs, - (CO)NR 7 R 8 , -SO 2 -NR 7 R 8 , -NR 7 SO 2 R 8 , -NR 7 R 8 , -OR 7 , -SR 7 , -0(CO)NR 7 R 8 , - NR 7 (CO)OR 8 , and -NR 7 SO 2 NR 8 , where R 7 and R 8 are independently selected from H or Z, where Z is selected from substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, polyalkylene glycol and polyols, where substituents thereon are selected from hydroxyls, amines, amidines, carboxylates, phosphonates, sulfonates, and guamdines, optionally linked to the ⁇ ng ArI by a heterocyclic linker, R 4 and R 12 are independently selected from H and R 7 , where R 7
  • each X is a halogen atom, which may be the same or different
  • Ri is selected from -SO 2 -NR 7 R 8 , -NR 7 (CO)R 8 , -(CO)NR 7 R 8 , -NR 7 SO 2 R 8 , -NR 7 R 8 , -OR 7 , -SR 7 , - 0(CO)NR 7 R 8 , -NR 7 (CO)OR 8 , and -NR 7 SO 2 NR 8 , where R 7 and R 8 are independently selected from H or Z, where Z is selected from substituted or unsubstituted hydrocarbyl,
  • R 3 is selected from H or R 7 , where R 7 is as desc ⁇ bed above, R 13 is selected from substituted or unsubstituted CpCg alkyl, R 2 and R ]2 are independently selected from H or R 7 , wherein R 7 is as desc ⁇ bed above, Rio and Ru, when present, are
  • ArI represents an aromatic nng, or alternatively a heteroaromatic ⁇ ng wherein one or more of the carbon atoms therein is replaced with a N, O or S atom
  • Ar2 represents an aromatic ⁇ ng, or alternatively a heteroaromatic ⁇ ng wherein one or more of the carbon atoms therein is replaced with a N, O or S atom
  • one of Ri, R 2 and R 3 is linked to the ⁇ ng ArI, and/or Rs is linked to the ⁇ ng Ar2, by a 5 heterocyclic linker having the structure
  • R represents Rj, R 2 , R 3 , or R 5 bound thereto
  • NHE Z molecule of the present disclosure may have the structure of Formula (IV)
  • each Ri, R 2 , R 3 , R 5 and R 9 are independently selected from H, halogen, NR 7 (CO)R 8 , -(CO)NR 7 R 8 , -SO 2 -NR 7 R 8 , -NR 7 SO 2 R 8 , -NR 7 R 8 , -OR 7 , -SR 7 , - 0(CO)NR 7 R 8 , -NR 7 (CO)OR 8 , and -NR 7 SO 2 NR 8 , where R 7 and R 8 are independently selected from H or Z, where Z is selected from substituted hydrocarbyl,
  • R 4 is selected from H or Z, where Z is substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, a polyalkylene glycol and a polyol, where substituents
  • R ⁇ is selected from -H and C 1 -C 7 alkyl
  • ArI and Ar2 independently represent an aromatic nng, or alternatively a heteroaromatic ⁇ ng wherein one or more of the carbon atoms therein is replaced with a N, O or S atom.
  • the compound may optionally have a tPSA of at least about 100 A 2 , about 150 A 2 , about 200 A 2 , about 250 A 2 , about 270 A 2 , or more and/or a molecular weight of at least about 710 Da
  • the compounds of the present disclosure comprise a NHE-mhibitmg small molecule that has been modified or functionahzed structurally to alter its physicochemical properties (by the attachment or inclusion of moiety Z), and more specifically the physicochemical properties of the NHE-Z molecule, thus rende ⁇ ng it substantially impermeable or substantially systemically non-bioavailable
  • the NHE-Z compound may be polyvalent (1 e , an oligomer, dendrimer or polymer moiety), wherein Z may be referred to in this embodiment generally as a "Core" moiety, and the NHE- inhibiting small molecule may be bound, directly or indirectly (by means of a linking moiety) thereto, the polyvalent compounds having for example one of the following general structures of Formula (VIII), (IX) and (X)
  • the NHE-inhibiting small molecule may be rendered substantially impermeable or substantially systemically non-bioavailable by forming a polymeric structure from multiple NHE-mhibiting small molecules, which may be the same or different, connected or bound by a se ⁇ es of linkers, L, which also may be the same or different, the compound having for example the structure of Formula (XI)
  • the polyvalent compound may be m dime ⁇ c, oligomenc or polymeric form, wherein for example Z or the Core is a backbone to which is bound (by means of a linker, for example) multiple NHE-inhibitmg small molecules
  • Such compounds may have, for example, the structures of Formulas (XIIA) or (XIIB)
  • NHE is a NHE-inhibitmg small molecule, each NHE as desc ⁇ bed above and in farther detail hereinafter, and n is a non-zero integer (i e , an integer of 1 or more)
  • the Core moiety has one or more attachment sites to which NHE- mhibiting small molecules are bound, and preferably covalently bound, via a bond or linker, L
  • the Core moiety may, m general, be anything that serves to enable the overall compound to be substantially impermeable or substantially systemically non- bioavailable (e g , an atom, a small molecule, etc ), but in one or more preferred embodiments is an oligomer, a dend ⁇ mer or a polymer moiety, in each case having more than one site of attachment for L (and thus for the NHE inhibiting small molecule)
  • the combination of the Core and NHE-mhibitmg small molecule i e , the "NHE-Z" molecule
  • repeat unit in Formulas (XIIA) and (XIIB) generally encompasses repeating units of va ⁇ ous polymeric embodiments, which may optionally be produced by methods referred to herein
  • each repeat unit may be the same or different, and may or may not be linked to the NHE-inhibiting small molecule by a linker, which in rum may be the same or different when present
  • linker which in rum may be the same or different when present
  • polyvalent refers to a molecule that has multiple (e g , 2, 4, 6, 8, 10 or more) NHE-inhibitmg moieties therein
  • certain polyvalent NHE-inhibiting compounds of the present disclosure show unexpectedly higher potency, as measured by inhibition assays (as farther detailed elsewhere herein) and charactenzed by the concentration of said NHE inhibitor resulting in 50% inhibition (l e , the IC 50 values)
  • certain multivalent structures represented generally by Formula (X), above, have an IC 50 value several fold lower in magnitude than the individual NHE, or L-NHE, structure (which may be referred to as the "monomer" or monovalent form)
  • multivalent compounds according to Formula (X) were observed to have an IC 50 value of at least about 5 time lower (1 e potency about 5 time higher) than the monomer (or monovalent) form (e g Examples 46 and 49)
  • multivalent compounds according to Formula (X) were observed to have an IC 50 value of at least about 10 time lower (1 e potency about 10 time higher) than the monomer form
  • the first representation below of an exemplary oligomer compound, wherein the various parts of the compound corresponding to the structure of Formula (X) are identified, is intended to provide a broad context for the disclosure provided herein
  • the linker moiety is a polyethylene glycol (PEG) motif
  • PEG de ⁇ vatives are advantageous due m part to their aqueous solubility, which may help avoid hydrophobic collapse (the intramolecular interaction of hydrophobic motifs that can occur when a hydrophobic molecule is exposed to an aqueous environment (see, e g , Wiley, R A , Rich, D H Medicai Research Reviews 1993, 13(3), 327-384)
  • the core moiety illustrated below is also advantageous because it provides some rigidity to
  • the structure may be for example
  • n and m may be independently selected from the range of from about 1 to about 10, more preferably from about 1 to about 5, and even more preferably from about 1 to about 2 In alternative embodiments, however, n and m may be independently selected from the range of from about 1 to about 500, preferably from about 1 to about 300, more preferably from about 1 to about 100, and most preferably from about 1 to about 50 In these or other particular embodiments, n and m may both be withm the range of from about 1 to about 50, or from about 1 to about 20
  • the structures provided above are illustrations of one embodiment of compounds utilized for administration wherein absorption is limited (i e , the compound is rendered substantially impermeable or substantially systemically non-bioavailable) by means of increasing the molecular weight of the NHE-inhibitmg small molecule
  • the NHE-inhibitmg small molecule may be rendered substantially impermeable or substantially systemically non- bioavailable by means of alte ⁇ ng, and more specifically increasing, the topological polar surface area, as further illustrated by the following structures, wherein a substituted aromatic rmg is bound to the "scaffold" of the NHE-inhibition small molecule
  • lomzable groups such as phosphonates, sulfonates, guamdmes and the like may be particularly advantageous at preventing paracellular permeability Carbohydates are also advantageous, and though uncharged, significantly increase tPSA while minimally increasing molecular weight
  • NHE-mhibiting small molecules suitable for use may, m particular, be selected independently from one or more of the small molecules desc ⁇ bed as benzoylguandines, heteroaroylguandines, "spacer-stretched” aroylguandmes, non-acyl guamdines and acylguanidine isosteres, above, and as discussed in further detail hereinafter and/or to the small molecules detailed in, for example US5866610, US6399824, US6911453, US6703405, US6005010, US6887870, US6737423, US7326705, US 55824691 (WO94/026709), US6399824 (WO02/024637), US 2004/0339001 (WO02/020496), US 2005
  • NHE-mhibiting compounds that may be utilized for the treatments detailed in the instant disclosure, it may in some cases be advantageous to first determine a likely point of attachment on a small molecule NHE inhibitor, where a core or linker might be installed or attached before making a senes of candidate multivalent or polyvalent compounds This may be done by one skilled in the art via known methods by systematically installing functional groups, or functional groups displaying a fragment of the desired core or linker, onto va ⁇ ous positions of the NHE inhibitor small molecule and then testing these adducts to determine whether the modified inhibitor still retains desired biological properties (e g , NHE inhibition)
  • An understanding of the SAR of the inhibitor also allows the design of cores and/or linkers that contribute positively to the activity of the resulting compounds For example, the SAR of an NHE inhibitor se ⁇ es may show that installation of an N-alkylated piperazme contributes positively to biochemical activity (increased potency) or pharmaceutical properties (increased so
  • Another aspect to be considered in the design of cores and linkers displaying an NHE inhibitor is the limiting or preventing of hydrophobic collapse Compounds with extended hydrocarbon functionalities may collapse upon themselves in an intramolecular fashion, causing an increased enthalpic barrier for interaction with the desired biological target
  • these are preferably designed to be resistant to hydrophobic collapse
  • conformational constraints such as ⁇ gid monocyclic, bicyclic or polycychc ⁇ ngs can be installed in a core or linker to increase the rigidity of the structure
  • Unsaturated bonds, such as alkenes and alkynes may also or alternatively be installed
  • Such modifications may ensure the NHE-inhibiting compound is accessible for productive binding with its target
  • the hydrophilicity of the linkers may be improved by adding hydrogen bond donor or acceptor motifs, or ionic motifs such as amines that are protonated in the GI, or acids that are deprotonated Such modifications will increase the hydrophilicity of the core or linker
  • NHE-inhibitmg small molecules modified consistent with the principles detailed above are illustrated below
  • These moieties display functional groups that facilitate their appendage to "Z" (e g , a core group, Core, or linking group, L)
  • These functional groups can include electrophiles, which can react with nucleophilic cores or linkers, and nucleophiles, which can react with electrophilic cores or linkers
  • Small molecule NHE inhibitors may be similarly de ⁇ vatized with, for example, boronic acid groups which can then react with approp ⁇ ate cores or linkers via palladium mediated cross-coupling reactions
  • the NHE inhibitor may also contain olefins which can then react with approp ⁇ ate cores or linkers via olefin metathesis chemistry, or alkynes or azides which can then react with approp ⁇ ate cores or linkers via [2 + 3] cycloaddtion
  • One skilled in the art may consider a va ⁇ ety of functional groups that will allow the facile
  • Cinnamoylguamdine NHE-inhibitmg Moiety Functionahzed to Display Electrophilic or Nucleophilic Groups to Facilitate Reaction with Cores and Linkers
  • Tetrahydroisoqumohne NHE-inhibiting Moiety Functionabzed to Display Electrophilic or Nucleophilic Groups to Facilitate Reaction with Cores and Linkers
  • va ⁇ ables in the above-noted structures are as defined m U S Patent No 6,911,453, the entire contents of which (and in particular the text of columns 1-4 therein) are incorporated herein by reference for all relevant and consistent purposes Scheme 3
  • va ⁇ ables in the above-noted structures are as defined m U S Patent Application No 2005/0020612 and U S Patent No 6,911,453, the entire contents of which (and in particular the text of columns 1-4 therein) are incorporated herein by reference for all relevant and consistent purposes
  • Exemplary electrophilic and nucleophilic linker moieties include, but are not limited to, the linker moieties illustrated in the Examples and the following:
  • R 2 -N 3 , -CO 2 H, -CHO, -OH, -SH,
  • the linking moiety, L, in each of the described embodiments can be a chemical linker, such as a bond or other moiety, for example, comprising about 1 to about 200 atoms, or about 1 to about 100 atoms, or about 1 to about 50 atoms, that can be hydrophilic and/or hydrophobic
  • the linking moiety can be a polymer moiety grafted onto a polymer backbone, for example, using living free radical polymerization approaches known in the art
  • Preferred L structures or moieties may also be selected from, for example, ohgoethylene glycol, oligopeptide, oligoethyleneimme, ohgotetramethylene glycol and oligocaprolactone
  • the core moiety can be an atom, a small molecule, an atom, a small molecule, an oligomer moiety, an oligomer moiety, or a nonrepeating moiety
  • the core moiety can be an atom, a small molecule, an atom, a small
  • Exemplary core moieties include but are not limited to the core moieties illustrated in the Examples and ether moieties, ester moieties, sulfide moieties, disulfide moieties, amine moieties, aryl moieties, alkoxyl moieties, etc , such as, for example, the following
  • each p, q, r and s is an independently selected integer ranging from about 0 to about 48, preferably from about 0 to about 36, or from about 0 to about 24, or from about 0 to about 16. In some instances, each p, q, r and s can be an independently selected integer ranging from about 0 to 12.
  • R can be a substituent moiety generally selected from halide, hydroxyl, amine, thiol, ether, carbonyl, carboxyl, ester, amide, carbocyclic, heterocyclic, and moieties comprising combinations thereof.
  • the core moiety is a dendrimer, defined as a repeatedly branched molecule (see, e.g., J. M. J. Frechet, D. A. Tomalia, Dendrimers and Other Dendritic Polymers, John Wiley & Sons, Ltd. NY, NY, 2001) and schematically represented below:
  • the NHE inhibiting small molecule is attached through L to one, several or optionally all termini located at the periphery of the dendrimer.
  • a dend ⁇ mer building block named dendron, and illustrated above is used as a core, wherein the NHE inhibitor group is attached to one, several or optionally all termini located at the periphery of the dendron.
  • the number of generations herein is typically between about 0 and about 6, and preferably between about 0 and about 3. (Generation is defined in, for example, J. M. J. Frechet, D. A. Tomalia, Dendrimers and Other Dendritic Polymers, John Wiley & Sons, Ltd.
  • Dendrimer and/or dendron structures are well known in the art and include, for example, those shown in or illustrated by: (i) J. M. J. Frechet, D. A. Tomalia, Dendrimers and Other Dendritic Polymers, John Wiley & Sons, Ltd NY, NY, ( ⁇ ) George R Newkome, Charles N Moorefield and F ⁇ tz Vogtle, Dendnmers and Dendrons Concepts, Syntheses, Applications, VCH Verlagsgesellschaft Mbh, and, (in) Boas, U , Ch ⁇ stensen, J B , Heegaard, P M H , Dendnmers in Medicine and Biotechnology New Molecular Tools , Springer, 2006
  • the core moiety may be a polymer moiety or an oligomer moiety
  • the polymer or oligomer may, in each case, be independently considered and compose repeat units consisting of a repeat moiety selected from alkyl ⁇ e g , -CH 2 -), substituted alkyl (e g , -CHR- , wherein, for example, R is hydroxy), alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, phenyl, aryl, heterocyclic, amine, ether, sulfide, disulfide, hydrazine, and any of the foregoing substituted with oxygen, sulfur, sulfonyl, phosphonyl, hydroxyl, alkoxyl, amine, thiol, ether, carbonyl, carboxyl, ester, amide, alkyl, alkenyl, alkynyl, aryl, heterocyclic, as well as moieties
  • Preferred polymers for polymeric moieties useful in constructing substantially impermeable or substantially systemically non-bioavailable NHE- inhibitmg compounds that are multivalent, for use in the treatment va ⁇ ous treatment methods disclosed herein can be prepared by any suitable technique, such as by free radical polymerization, condensation polymerization, addition polymerization, ring- opening polymerization, and/or can be derived from naturally occurring polymers, such as saccharide polymers Further, in some embodiments, any of these polymer moieties may be functionahzed Examples of polysaccha ⁇ des useful in preparation of such compounds include but are not limited to mate ⁇ als from vegetable or animal origin, including cellulose mate ⁇ als, hemicellulose, alkyl cellulose, hydroxyalkyl cellulose, carboxymethylcellulose, sulfoethylcellulose, starch, xylan, amylopectine, chondroitm, hyarulonate, heparin, guar, xanthan, mannan, galactomannan, chi
  • the polymer moiety can be prepared from va ⁇ ous classes of monomers including, for example, acrylic, methacrylic, styremc, vinylic, and dienic, whose typical examples are given thereafter styrene, substituted styrene, alkyl acrylate, substituted alkyl acrylate, alkyl methacrylate, substituted alkyl methacrylate, acrylomt ⁇ le, methacrylomtnle, acrylamide, methacrylamide, N-alkylacrylamide, N-alkylmethacrylamide, N,N- dialkylacrylamide, N,N-dialkylmethacrylamide, isoprene, butadiene, ethylene, vmyl acetate, and combinations thereof Functionahzed versions of these monomers may also be used and any of these monomers may be used with other monomers as comonomers
  • specific monomers or comonomers that may be used in this disclosure include methyl
  • the polymer to which the NHE inhibitor small molecule, NHE, is attached or otherwise a part of is a polyol (e g , a polymer having a repeat unit of, for example, a hydroxyl-substituted alkyl, such as -CH(OH)-)
  • Polyols such as mono- and disaccharides, with or without reducing or reducible end groups thereon, may be good candidates, for example, for installing additional functionality that could render the compound substantially impermeable
  • the NHE inhibiting small molecule is attached at one or both ends of the polymer chain
  • a macromolecule e g , a polymer or oligomer having one of the following exemplary structures may be designed and constructed as desc ⁇ bed herein
  • repeat moiety m Formulas (XIIA) or (XIIB) generally encompasses repeating units of polymers and copolymers produced by methods referred to herein above
  • the various properties of the oligomers and polymers that form the core moiety as disclosed herein above may be optimized for a given use or application using experimental means and principles generally known m the art
  • the overall molecular weight of the compounds or structures presented herein above may be selected so as to achieve non-absorbability, inhibition persistence and/or potency
  • the polymer moiety is stable under physiological conditions of the GI tract
  • stable it is meant that the polymer moiety does not degrade or does not degrade significantly or essentially does not degrade under the physiological conditions of the GI tract
  • at least about 90%, preferably at least about 95%, and more preferably at least about 98%, and even more preferably at least about 99% of the polymer moiety remains un-degraded or intact after at least about 5 hours, at least about 12 hours, at least about 18 hours, at least about 24 hours, or at least about 48 hours of residence in a gastrointestinal tract
  • Stability in a gastrointestinal tract can be evaluated using gastrointestinal mimics, e g , gastric mimics or intestinal mimics of the small intestine, which approximately model the physiological conditions at one or
  • modifications preferably include addition of di- anions, such as phosphonates, malonates, sulfonates and the like, and polyols such as carbohydrates and the like
  • di- anions such as phosphonates, malonates, sulfonates and the like
  • polyols such as carbohydrates and the like
  • Exemplary derivatives of NHEs with increased tPSA include but are not limited to the following
  • the "NHE-Z" molecule is polyvalent, that is, the molecule contains two or
  • the NHE-Z molecule may be selected, for example, from one of the following Formulas (IV), (V), (VI) or (VII)
  • each Ri, R 2 , R 3 , R 5 and R 9 are independently selected from H, halogen, - NR 7 (CO)R 8 , -(CO)NR 7 R 8 , -SO 2 -NR 7 R 8 , -NR 7 SO 2 R 8 , -NR 7 R 8 , -OR 7 , -SR 7 , - 0(CO)NR 7 R 8 , -NR 7 (CO)OR 8 , and -NR 7 SO 2 NR 8 , where R 7 and R 8 are independently selected from H or L, provided at least one is L, wherein L is selected from the group consisting of substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, polyalkylene glycol and polyols, and further wherein L links the repeat unit to at least one other repeat unit and/or at least one other Core moiety independently selected from substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, polyalkylene glycol, polyols
  • each Ri, R 2 , R 3 , and R 5 are optionally linked to the ⁇ ng ArI by a heterocyclic linker, and further are independently selected from H, -NR 7 (CO)R 8 , -(CO)NR 7 R 8 , -SO 2 - NR 7 R 5 , NR 7 SO 2 R 8 , -NR 7 R 8 , -OR 7 , -SR 7 , -0(CO)NR 7 R 8 , -NR 7 (CO)OR 8 , and - NR 7 SO 2 NR 8 , where R 7 and R 8 are independently selected from H or L, provided at least one is L, wherein L is selected from the group consisting of substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, polyalkylene glycol and polyols, and further wherein L links the repeat unit to at least one other repeat unit and/or at least one other Core moiety independently selected from substituted or unsubstituted hydrocarbyl, heterohydro
  • each X is a halogen atom, which may be the same or different
  • Ri is selected from -SO 2 -NR 7 R 8 , -NR 7 (CO)R 8 , -(CO)NR 7 R 8 , -NR 7 SO 2 R 8 , -NR 7 R 8 , -OR 7 , -SR 7 , - 0(CO)NR 7 R 8 , -NR 7 (CO)OR 8 , and -NR 7 SO 2 NR 8 , where R 7 and R 8 are independently selected from H or L, provided at least one is L, wherein L is selected from the group consisting of substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, polyalkylene glycol and polyols, and further wherein L links the repeat unit to at least one other repeat unit and/or at least one other Core moiety independently selected from substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, polyalkylene glycol, polyols, polyamine
  • one of Ri, R 2 and R 3 is linked to the ⁇ ng ArI, and/or R 5 is linked to the ⁇ ng Ar2, by a heterocyclic linker having the structure
  • NHE-inhibitmg small molecule has the structure of Formula (IV)
  • each Ri, R 2 , R 3 , R 5 and R 9 are independently selected from H, halogen, -NRv(CO)Rs, - (CO)NR 7 R 8 , -SO 2 -NR 7 R 8 , -NR 7 SO 2 R 8 , -NR 7 R 8 , -OR 7 , -SR 7 , -0(CO)NR 7 R 8 , - NR 7 (CO)OR 8 , and -NR 7 SO 2 NR 8 , where R 7 and R 8 are independently selected from H or a bond linking the NHE-inhibiting small molecule to L, provided at least one is a bond linking the NHE-inhibitmg small molecule to L, R 4 is selected from H, Ci-C 7 alkyl, or a bond linking the NHE-inhibitmg small molecule to L, Ke is absent or selected from H and C 1 -C 7 alkyl, and
  • the NHE- mhibiting small molecule has the following structure
  • each Ri, R 2 and R 3 are independently selected from H, halogen, -NR 7 (CO)R 8 , -(CO)NR 7 R 8 , - SO 2 -NR 7 R 8 , NR 7 SO 2 R 8 , -NR 7 R 8 , -OR 7 , -SR 7 , -0(CO)NR 7 R 8 , -NR 7 (CO)OR 8 , and - NR 7 SO 2 NR 8 , where R 7 and R 8 are independently selected from H or a bond linking the NHE-inhibitmg small molecule to L, provided at least one is a bond linking the NHE- inhibiting small molecule to L
  • the NHE- mhibitmg small molecule has one of the following structures
  • L is a polyalkylene glycol linker, such as a polyethylene glycol linker
  • n 2
  • the Core is selected from the group consisting of
  • Terminology Physical and Performance Properties
  • Niro refers to the -NO2 radical
  • Alkyl refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, which is saturated or unsaturated (; e , contains one or more double and/or triple bonds), having from one to twelve carbon atoms (C 1 -C 12 alkyl), preferably one to eight carbon atoms (Ci-Cg alkyl) or one to six carbon atoms (C 1 -C 6 alkyl), and which is attached to the rest of the molecule by a single bond, e g , methyl, ethyl, rc-propyl, 1-methylethyl (wo-propyl), rc-butyl, n-pentyl,
  • alkyl group may be optionally substituted
  • Alkylene or “alkylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, which is saturated or unsaturated (1 e , contains one or more double and/or triple bonds), and having from one to twelve carbon atoms, e g , methylene, ethylene, propylene, «-butylene, ethenylene, propenylene, «-butenylene, propynylene, rc-butynylene, and the like
  • the alkylene chain is attached to the rest of the molecule through a single or double bond and to the radical group through a single or double bond
  • the points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain Unless stated otherwise specifically in the specification, an alkylene chain may be optionally substituted "Alkoxy" refers to a radical of the formula -OR a
  • Thioalkyl refers to a radical of the formula -SR a where R a is an alkyl radical as defined above containing one to twelve carbon atoms Unless stated otherwise specifically in the specification, a thioalkyl group may be optionally substituted
  • Aryl refers to a hydrocarbon nng system radical comp ⁇ sing hydrogen, 6 to 18 carbon atoms and at least one aromatic nng
  • the aryl radical may be a monocyclic, bicyclic, t ⁇ cyclic or tetracyclic nng system, which may include fused or bndged nng systems
  • Aryl radicals include, but are not limited to, aryl radicals denved from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, fluoranthene, fluorene, as indacene, 5-indacene, mdane, mdene, naphthalene, phenalene, phenanthrene, pleiadene, pyrene, and tnphenylene Unless stated otherwise specifically in the specification, the term "aryl” or the prefix "ar-"
  • Aralkyl refers to a radical of the formula -R b -R 0 where R b is an alkylene chain as defined above and R 0 is one or more aryl radicals as defined above, for example benzyl, diphenylmethyl and the like Unless stated otherwise specifically in the specification, an aralkyl group may be optionally substituted
  • Cycloalkyl or “carbocyclic nng” refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, which may include fused or bndged nng systems, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond
  • Monocyclic radicals include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl
  • Polycyclic radicals include, for example, adamantyl, norbornyl, decalmyl, 7,7-dimethyl-bicyclo[2 2 ljheptanyl, and the like Unless otherwise stated specifically in the specification, a cycloalkyl group may be optionally substituted
  • Cycloalkylalkyl refers to a radical of the formula -R b R d where Ra is an alkylene chain as defined above and R g is a cycloalkyl radical as defined above Unless stated otherwise specifically in the specification, a cycloalkylalkyl group may be optionally substituted "Fused” refers to any ring structure described herein which is fused to an existing ⁇ ng structure in the compounds of the invention When the fused ⁇ ng is a heterocyclyl ⁇ ng or a heteroaryl ⁇ ng, any carbon atom on the existing ⁇ ng structure which becomes part of the fused heterocyclyl ⁇ ng or the fused heteroaryl ⁇ ng may be replaced with a nitrogen atom "Halo" or “halogen” refers to bromo, chloro, fluoro or iodo
  • Haloalkyl refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e g , t ⁇ fluoromethyl, difluoromethyl, tnchloromethyl, 2,2,2-t ⁇ fluoroethyl, 1 ,2-difluoroethyl,
  • haloalkyl group may be optionally substituted
  • Heterocyclyl or “heterocyclic ⁇ ng” refers to a stable 3- to 18-membered non-aromatic ⁇ ng radical which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur
  • the heterocyclyl radical may be a monocyclic, bicychc, tricyclic or tetracyclic ⁇ ng system, which may include fused or b ⁇ dged ⁇ ng systems
  • the nitrogen, carbon or sulfur atoms in the heterocyclyl radical may be optionally oxidized, the nitrogen atom may be optionally quaternized, and the heterocyclyl radical may be partially or fully saturated
  • Examples of such heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl[l,3]dithianyl, decahydroisoqumolyl, lmidazohnyl, lmidazolidmyl, lso
  • iV-heterocyclyl refers to a heterocyclyl radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a nitrogen atom in the heterocyclyl radical Unless stated otherwise specifically in the specification, a iV-heterocyclyl group may be optionally substituted
  • Heterocyclylalkyl refers to a radical of the formula -R b R e where R b is an alkylene chain as defined above and R e is a heterocyclyl radical as defined above, and if the heterocyclyl is a mtrogen-contaimng heterocyclyl, the heterocyclyl may be attached to the alkyl radical at the nitrogen atom Unless stated otherwise specifically m the specification, a heterocyclylalkyl group may be optionally substituted
  • Heteroaryl refers to a 5- to 14-membered ⁇ ng system radical comp ⁇ sing hydrogen atoms, one to thirteen carbon atoms, one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and at least one aromatic ⁇ ng
  • the heteroaryl radical may be a monocyclic, bicychc, tricyclic or tetracyclic ⁇ ng system, which may include fused or b ⁇ dged nng systems
  • the nitrogen, carbon or sulfur atoms in the heteroaryl radical may be optionally oxidized, the nitrogen atom may be optionally quatermzed Examples include, but are not limited to, azepinyl, ac ⁇ dinyl, benzimidazolyl, benzothiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[6][l,4
  • 'W-heteroaryl refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical Unless stated otherwise specifically in the specification, an JV-heteroaryl group may be optionally substituted
  • Heteroarylalkyl refers to a radical of the formula -R b R f where R b is an alkylene chain as defined above and R f is a heteroaryl radical as defined above Unless stated otherwise specifically in the specification, a heteroarylalkyl group may be optionally substituted
  • substituted means any of the above groups (z e , alkyl, alkylene, alkoxy, alkylammo, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, iV-heterocyclyl, heterocyclylalkyl, heteroaryl, ⁇ '-heteroaryl and/or heteroarylalkyl) wherein at least one hydrogen atom is replaced by a bond to a non-hydrogen atoms such as, but not limited to a halogen atom such as F, Cl, Br, and I, an oxygen atom in groups such as hydroxyl groups, alkoxy groups, and ester groups, a sulfur atom in groups such as thiol groups, thioalkyl groups, sulfone groups, sulfonyl groups, and sulfoxide groups, a nitrogen atom in
  • Prodrug is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound of the invention
  • prodrug refers to a metabolic precursor of a compound of the invention that is pharmaceutically acceptable
  • a prodrug may be inactive when administered to a subject in need thereof, but is converted in vivo to an active compound of the invention
  • Prodrugs are typically rapidly transformed in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood
  • the prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, Bundgard, H , Design of Prodrugs (1985), pp 7-9, 21-24 (Elsevier, Amsterdam))
  • a discussion of prodrugs is provided in Higuchi, T , et al , A C S Symposium Se ⁇ es, VoI 14, and in Bioreversible Carriers in Drug Design, Ed Edward B Roche, American Pharmaceutical Association and Pergamon Press, 1987
  • prodrug is also
  • the invention disclosed herein is also meant to encompass the in vivo metabolic products of the disclosed compounds Such products may result from, for example, the oxidation, reduction, hydrolysis, amidation, estenfication, and the like of the administered compound, primarily due to enzymatic processes
  • the invention includes compounds produced by a process composing administering a compound of this invention to a mammal for a pe ⁇ od of time sufficient to yield a metabolic product thereof
  • Such products are typically identified by administering a radiolabeled compound of the invention in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to human, allowing sufficient time for metabolism to occur, and isolating its conversion products from the u ⁇ ne, blood or other biological samples
  • “Stable compound” and “stable structure” are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of pu ⁇ ty from a reaction mixture, and formulation into an efficacious therapeutic agent
  • “Optional” or “optionally” means that the subsequently desc ⁇ bed event or circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not
  • “optionally substituted aryl” means that the aryl radical may or may not be substituted and that the desc ⁇ ption includes both substituted aryl radicals and aryl radicals having no substitution
  • “Pharmaceutically acceptable earner, diluent or excipient” includes without limitation any adjuvant, earner, excipient, ghdant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved
  • “Pharmaceutically acceptable salt” includes both acid and base addition salts
  • “Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but are not limited to, hydrochlonc acid, hydrobromic acid, sulfuric acid, nitric acid, phosphonc acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfomc acid, benzoic acid, 4-acetamidobenzoic acid, camphonc acid, camphor-10-sulfonic acid, cap ⁇ c acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citnc acid, cyclamic acid, dodecylsulfunc acid, ethane- 1 ,2-disulfomc acid, ethanesulfomc
  • “Pharmaceutically acceptable base addition salt” refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable
  • These salts are prepared from addition of an inorganic base or an organic base to the free acid
  • Salts denved from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zmc, copper, manganese, aluminum salts and the like
  • Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts
  • Salts de ⁇ ved from organic bases include, but are not limited to, salts of primary, S secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, lsopropylamme, t ⁇ methylamme, diethylaimne, t ⁇ ethylamme, t ⁇ propylamme
  • Particularly preferred organic bases are lsopropylamme, diethylamme, ethanolamme, t ⁇ methylamine, dicyclohexylamine, choline and caffeine
  • solvate refers to an aggregate that comp ⁇ ses one or more molecules of a compound of the invention with one or more molecules of solvent
  • the solvent may be water, in which case the solvate may be a hydrate Alternatively, the solvent may be an organic solvent
  • the compounds of the invention may be water, in which case the solvate may be a hydrate Alternatively, the solvent may be an organic solvent
  • the 20 present invention may exist as a hydrate, including a monohydrate, dihydrate, hermhydrate, sesquihydrate, tnhydrate, tetrahydrate and the like, as well as the corresponding solvated forms
  • the compound of the invention may be true solvates, while in other cases, the compound of the invention may merely retain adventitious water or be a mixture of water plus some adventitious solvent
  • a “pharmaceutical composition” refers to a formulation of a compound of the invention and a medium generally accepted m the art for the delivery of the biologically active compound to mammals, e g , humans Such a medium includes all pharmaceutically acceptable earners, diluents or excipients therefor
  • 30 salts may contain one or more asymmetric centers and may thus give ⁇ se to enantiomers, diastereomers, and other stereoisome ⁇ c forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for ammo acids
  • the present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms Optically active (+) and (-), (R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization
  • Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or de ⁇ vative) using, for example, chiral high pressure liquid chromatography (HPLC) When the compounds desc
  • a “stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable
  • the present invention contemplates va ⁇ ous stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molecules are nonsupe ⁇ mposeable mirror images of one another
  • a “tautomer” refers to a proton shift from one atom of a molecule to another atom of the same molecule
  • the present invention includes tautomers of any said compounds
  • the compounds descnbed herein are designed to be substantially active or localized in the gastrointestinal lumen of a human or animal subject
  • gastrointestinal lumen is used interchangeably herein with the term “lumen,” to refer to the space or cavity within a gastrointestinal tract (GI tract, which can also be referred to as the gut), delimited by the apical membrane of GI epithelial cells of the subject
  • the compounds are not absorbed through the layer of epithelial cells of the GI tract (also known as the GI epithelium)
  • Gastrointestinal mucosa refers to the layer(s) of cells separating the gastrointestinal lumen from the rest of the body and includes gastric and intestinal mucosa, such as the mucosa of the small intestine
  • a "gastrointestinal epithelial cell” or a “gut epithelial cell” as used herein refers to any epithelial cell on the surface of the gastrointestinal mucosa that faces the lumen of the gastrointestinal tract
  • Substantially systemically non-bioavailable and/or “substantially 5 impermeable” as used herein (as well as va ⁇ ations thereof) generally refer to situations in which a statistically significant amount, and in some embodiments essentially all of the compound of the present disclosure (which includes the NHE-inhibitor small molecule), remains in the gastrointestinal lumen
  • m accordance with one or more embodiments of the present disclosure, preferably at least about 70%, about
  • localization to the gastrointestinal lumen refers to reducing net movement across a gastrointestinal layer of epithelial cells, for example, by way of both transcellular and paracellular transport, as well as by active and/or passive transport
  • the compound m such embodiments is
  • I 5 hindered from net permeation of a layer of gastrointestinal epithelial cells in transcellular transport, for example, through an apical membrane of an epithelial cell of the small intestine
  • the compound in these embodiments is also hindered from net permeation through the "tight junctions" in paracellular transport between gastrointestinal epithelial cells lining the lumen
  • the compound is essentially not absorbed at all by the GI tract or gastrointestinal lumen
  • the terms “substantially impermeable” or “substantially systemically non- bioavailable” refers to embodiments wherein no detectable amount of absorption or permeation or systemic exposure of the compound is detected, using means generally
  • substantially impermeable or “substantially systemically non bioavailable” provides or allows for some limited absorption in the GI tract, and more particularly the gut epithelium, to occur (e g , some detectable amount of absorption,
  • substantially impermeable or “substantially systemically non bioavailable” refers to compounds that exhibit some detectable permeability to an epithelium layer of cells in the GI tract of less than about 20% of the administered compound (e g , less than about 15%, about 10%, or even about 5%, and for example greater than about 0 5%, or 1%), but then are cleared by the liver (i e , hepatic extraction) and/or the kidney (i e , renal excretion)
  • the ability of the compound to be substantially systemically non-bioavailable is based on the compound charge, size, and/or other physicochemical parameters (e g , polar surface area, number of hydrogen bond donors and/or acceptors therein, number of freely rotatable bonds, etc ) More specifically, it is to be noted that the absorption character of a compound can be selected by applying principles of pharmacodynamics, for example, by applying Lipinski's rule, also known as "the rule of five " Although not a rule, but rather a set of guidelines, Lipmski shows that small molecule drugs with (i) a molecular weight, (ii) a number of hydrogen bond donors, (in) a number of hydrogen bond acceptors, and/or (iv) a water/octanol partition coefficient (Mo ⁇ guchi Log P), greater than a certain threshold value, generally do not show significant systemic concentration (i e , are generally not absorbed to any
  • the molecular polar surface area (i e , "PSA"), which may be characterized as the surface belonging to polar atoms, is a desc ⁇ ptor that has also been shown to correlate
  • PSA is expressed in A 2 (squared angstroms) and is computed from a three dimensional molecular representation
  • a fast calculation method is now available (see, e g , Ertl et al , Journal of Medicinal Chemistry, 2000, 43,
  • topological PSA topological PSA
  • tPSA topological PSA
  • the compounds of the present disclosure may be constructed to exhibit a tPSA value greater than about 100 A 2 , about 120 A 2 , about 130 A 2 , or about 140 A 2 , and in some instances about 150 A 2 , about 200 A 2 , about 250 A 2 , about 270 A 2 , about 300 A 2 , about 400 A 2 ,or even about 500 A 2 , such that the compounds are substantially impermeable or substantially systemically non-bioavailable (as defined elsewhere herein).
  • the permeability properties of the compounds of the present disclosure may be screened experimentally.
  • the permeability coefficient can be determined by methods known to those of skill in the art, including for example by Caco-2 cell permeability assay and/or using an artificial membrane as a model of a gastrointestinal epithelial cell. (As previously noted above, see for example U.S. Patent No. 6,737,423, Example 31 for a description of the Caco-2 Model, which is incorporated herein by reference).
  • a synthetic membrane impregnated with, for example, lecithin and/or dodecane to mimic the net permeability characteristics of a gastrointestinal mucosa may be utilized as a model of a gastrointestinal mucosa.
  • the membrane can be used to separate a compartment containing the compound of the present disclosure from a compartment where the rate of permeation will be monitored.
  • the compounds utilized in the methods of the present disclosure may have a permeability coefficient, P app , of less than about 100 x 10 6 cm/s, or less than about 10 x 10 6 cm/s, or less than about 1 x 10 6 cm/s, or less than about 0 1 x 10 6 cm/s, when measured using means known in the art (such as for example the permeability experiment desc ⁇ bed m Whysland et al , J Med Chem , 2001, 44 923-930, the contents of which is incorporated here
  • NHE inhibitor small molecules are modified as desc ⁇ bed above to hinder the net absorption through a layer of gut epithelial cells, rende ⁇ ng them substantially systemically non- bioavailable
  • the compounds of the present disclosure comp ⁇ se an NHE-mhibiting small molecule linked, coupled or otherwise attached to a moiety Z, which may be an oligomer moiety, a polymer moiety, a hydrophobic moiety, a hydrophilic moiety, and/or a charged moiety, which renders the overall compound substantially impermeable or substantially systemically non bioavailable
  • the NHE-inhibiting small molecule is coupled to a multimer or polymer portion or moiety, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non bioavailable
  • the multimer or polymer portion or moiety may be of a molecular weight greater than about 500 Daltons (Da), about 1000 Da, about 2500 Da, about 5000 Da, about 10,000
  • the substantially impermeable or substantially systemically non-bioavailable NHE-mhibitmg compounds utilized in the treatment methods of the present disclosure may additionally exhibit a persistent inhibitor effect
  • This effect manifests itself when the inhibitory action of a compound at a certain concentration in equilibrium with the epithelial cell (e g , at or above its inhibitory concentration, IC) does not revert to baseline (i e , sodium transport without inhibitor) after the compound is depleted by simple washing of the luminal content
  • IC inhibitory concentration in equilibrium with the epithelial cell
  • the persistence effect can be determined using in vitro methods, in one instance, cell lines expressing NHE transporters are split in different vials and treated with a NHE inhibiting compound and sodium solution to measure the rate of sodium uptake The cells m one set of vials are washed for different penods of time to remove the inhibitor, and sodium uptake measurement is repeated after the washing Compounds that maintain their inhibitory effect after multiple/lengthy washing steps (compared to the inhibitory effect measured in the vials where washing does not occur) are persistent inhibitors Persistence effect can also be characterized ex vivo by using the everted sac technique, whereby transport of Na is monitored using an excised segment of GI perfused with a solution containing the inhibitor and shortly after flushing the bathing solution with a buffer solution free from inhibitor A persistence effect can also be characte ⁇ zed in vivo by observing the time needed for sodium balance to return to normal when the inhibitor treatment is discontinued The limit of the method resides in the fact that apical cells (and therefore apical NHE transporters
  • compounds of the present disclosure are resistant to enzymatic metabolism
  • administered compounds are preferably resistant to the activity of P450 enzymes, glucurosyl transferases, sulfotransferases, glutathione S-transferases, and the like, in the intestinal mucosa, as well as gast ⁇ c (e g , gastric lipase, and pepsme), pancreatic (e g , trypsin, triglyceride pancreatic lipase, phosphohpase A2, endonucleases, nucleotidases, and alpha-amylase), and brush-border enzymes (e g , alkaline phosphatase, glycos
  • the compounds that are utilized in methods of the present disclosure are also preferably resistant to metabolism by the bacte ⁇ al flora of the gut, that is, the compounds are not substrates for enzymes produced by bacte ⁇ al flora
  • the compounds administered in accordance with the methods of the present disclosure may be substantially inactive towards the gastrointestinal flora, and do not disrupt bacte ⁇ al growth or survival
  • the minimal inhibitory concentration (or "MIC") against GI flora is desirably greater than about 15 ⁇ g/ml, about 30 ⁇ g/ml, about 60 ⁇ g/ml, about 120 ⁇ g/ml, or even about 240 ⁇ g/ml, the MIC in vanous embodiments being for example between about 16 and about 32 ⁇ g/ml, or between about 64 and about 128 ⁇ g/ml, or greater than about 256 ⁇ g/ml
  • metabolic stability can be achieved in a number of ways Functionality susceptible to P450-mediated oxidation can be protected by, for example, blocking the point of metabolism with a halogen or other functional group Alternatively, electron withdrawing groups can be added to a conjugated system to generally provide protection to oxidation by reducing the electrophilicity of the compound Proteolytic stability can be achieved by avoiding secondary amide bonds, or by incorporating changes in stereochemistry or other modifications that prevent the drug from otherwise being recognized as a substrate by the metabolizing enzyme
  • one or more of the NHE-Z inhibiting compounds (monovalent or divalent) detailed herein when administered either alone or in combination with one or more additional pharmaceutically active compounds or agents (including, for example, a fluid-absorbing polymer) to a patient in need thereof, may act to increase the patient's daily fecal output of sodium by at least about 20, about 30 mmol, about 40 mmol, about 50 mmol, about 60 mmol, about 70 mmol, about 80 mmol, about 90 mmol, about 100 mmol, about 125 mmol, about 150 mmol or more, the increase being for example within the range of from about 20 to about 150 mmol/day, or from about 25 to about 100 mmol/day, or from about 30 to about 60 mmol/day
  • one or more of the NHE-Z inhibiting compounds (monovalent or divalent) detailed herein when administered either alone or in combination with one or more additional pharmaceutically active compounds or agents (including, for example, a fluid-absorbing polymer) to a patent in need thereof, may act to increase the patient's daily fluid output by at least about 100 ml, about 200 ml, about 300 ml, about 400 ml, about 500 ml, about 600 ml, about 700 ml, about 800 ml, about 900 ml, about 1000 ml or more, the increase being for example within the range of from about 100 to about 1000 ml/day, or from about 150 to about 750 ml/day, or from about 200 to about 500 ml/day (assuming isotonic fluid)
  • NHE-Z inhibiting compounds monovalent or divalent
  • additional pharmaceutically active compounds or agents including, for example, a fluid-absorbing polymer
  • 10% increase in fecal water content has a C raax that is less than the IC 50 for NHE-3, more specifically, less than about 1OX (10 times) the IC 50 , and, more specifically still, less than about IOOX (100 times) the IC 50
  • one or more of the NHE-Z inhibiting compounds (monovalent or divalent) detailed herein when administered either alone or in combination with one or more additional pharmaceutically active compounds or agents (including, for example, a fluid-absorbing polymer) to a patient in need thereof, may have a C max of less than about 10 ng/ml, about 7 5 ng/ml, about 5 ng/ml, about 2 5 ng/ml, about 1 ng/ml, or about 0 5 ng/ml, the C m8x bemg for example within the range of about 1 ng/ml to about 10 ng/ml, or about 2 5 ng/ml to about 7 5 ng/ml
  • one or more of the NHE-Z inhibiting compounds (monovalent or divalent) detailed herein when administered either alone or in combination with one or more additional pharmaceutically active compounds or agents (including, for example, a fluid-absorbing polymer) to a patient in need thereof, may have a IC 5O of less than about 10 ⁇ M, about 7 5 ⁇ M, about 5 ⁇ M, about 2 5 ⁇ M, about 1 ⁇ M, or about 0 5 ⁇ M, the IC 50 being for example withm the range of about 1 ⁇ M to about 10 ⁇ M, or about 2 5 ⁇ M to about 7 5 ⁇ M
  • one or more of the NHE-Z inhibiting compounds (monovalent or divalent) detailed herein when administered to a patient in need thereof, may have a ratio of IC 50 C max , wherein IC 50 and C max are expressed in terms of the same units, of at least about 10, about 50, about 100, about 250, about 500, about 750, or about 1000
  • the maximum compound concentration detected m the serum is lower than the NHE inhibitory concentration IC 50 of said compound
  • IC 50 is defined as the quantitative measure indicating the concentration of the compound required to inhibit 50% of the NHE-mediated Na / H antiport activity in a cell based assay
  • a pharmaceutical composition or preparation that may be used in accordance with the present disclosure for the treatment of various disorders associated with fluid retention and/or salt overload in the gastrointestinal tract (e g , hypertension, heart failure (in particular, congestive heart failure), chrome kidney disease, end-stage renal disease, liver disease and/or peroxisome prohferator-activated receptor (PPAR) gamma agonist-induced fluid retention) comp ⁇ ses, in general, the substantially impermeable or substantially systemically non-bioavailable NHE-inhibiting compound of the present disclosure, as well as various other optional components as further detailed herein below (e g , pharmaceutically acceptable excipients, etc )
  • the compounds utilized in the treatment methods of the present disclosure, as well as the pharmaceutical compositions comprising them may accordingly be administered alone, or as part of a treatment protocol or regiment that includes the administration or use of other beneficial compounds (as further detailed elsewhere herein)
  • the NHE-inhibitmg may accordingly be administered alone, or as part of a treatment protocol or regiment that includes the administration or
  • a “subject” or “mammal” is preferably a human, but can also be an animal in need of treatment with a compound of the disclosure, e g , companion animals (e g , dogs, cats, and the like), farm animals (e g , cows, pigs, horses and the like) and laboratory animals (e g , rats, mice, guinea pigs and the like)
  • Subjects "in need of treatment” with a compound of the present disclosure, or subjects “in need of NHE inhibition” include subjects with diseases and/or conditions that can be treated with substantially impermeable or substantially systemically non-bioavailable NHE-inhibitmg compounds, with or without a fluid- absorbing polymer, to achieve a beneficial therapeutic and/or prophylactic result
  • a beneficial outcome includes a decrease in the seventy of symptoms or delay in the onset of symptoms, increased longevity and/or more rapid or more complete resolution of the disease or condition
  • a subject in need of treatment may be suffering from hypertension, from salt-sensitive hypertension which may result from dietary salt intake, from a ⁇ sk of a cardiovascular disorder (e g , myocardial infarction, congestive heart failure and the like) resulting from hypertension, from heart failure (e g , congestive heart failure) resulting m fluid or salt overload, from chronic kidney disease resulting m fluid or salt overload, from end stage renal disease resulting in fluid or salt overload, from liver disease
  • NHE inhibitors may be beneficial for patients put on "non added salt” dietary regimen (i e , 60-100 mmol of Na per day), to liberalize their diet while keeping a neutral or slightly negative sodium balance (i e , the overall uptake of salt would be equal of less than the secreted salt)
  • “liberalize their diet” means that patients treated may add salt to their meals to make the meals more palatable, or/and diversify their diet with salt-containing foods, thus maintaining a good nutntional status while improving their quality of life
  • the treatment methods desc ⁇ bed herein may also help patients with edema associated with chemotherapy, pre menstrual fluid overload and preeclampsia (pregnancy-induced hypertension)
  • the present disclosure is further directed to methods of treatment involving the administration of the compound of the present disclosure, or a pharmaceutical composition comprising such a compound
  • Such methods may include, for example, a method for treating hypertension, the method comprising administering to the patient a substantially impermeable or substantially systemically non-bioavailable NHE-inhibiting compound, or a composition compnsing it
  • the method may be for reducing fluid overload associated with heart failure (in particular, congestive heart failure), the method comprising administering to the patient a substantially impermeable or substantially systemically 5 non-bioavailable NHE-inhibiting compound or pharmaceutical composition compnsing it
  • the method may be for reducing fluid overload associated with end stage renal disease, the method compnsing administenng to the patient a substantially impermeable or substantially systemically non-bioavailable NHE-inhibitmg compound or composition compnsing it
  • the method may be for reducing fluid overload associated
  • the method compnsing administenng to the patient a substantially impermeable or substantially systemically non-bioavailable NHE-inhibiting compound or composition compnsing it Additionally, or alternatively, the method may be for decreasing the activity of an intestinal NHE transporter in a patient, the method compnsmg
  • PPAR peroxisome proliferator-activated receptor
  • a pharmaceutical composition or preparation that may be used m
  • vanous gastrointestinal tract disorders comprising, in general, any small molecule, which may be monovalent or polyvalent, that is effective or active as an NHE-inhibitor and that is substantially active in the GI tract, in particular, the substantially impermeable or substantially systemically
  • NHE-inhibiting compound of the present disclosure 2 5 non-bioavailable NHE-inhibiting compound of the present disclosure, as well as vanous other optional components as further detailed herein below (e g , pharmaceutically acceptable excipients, etc )
  • the compounds utilized in the treatment methods of the present disclosure, as well as the pharmaceutical compositions compnsing them, may accordingly be administered alone, or as part of a treatment protocol or regiment that
  • the NHE-inhibitmg compound including any pharmaceutical composition composing the compound, is administered with a fluid-absorbing polymer (as more fully desc ⁇ bed below)
  • a "subject” is preferably a human, but can also be an animal m need of treatment with a compound of the disclosure, e g , companion animals (e g , dogs, cats, and the like), farm animals (e g , cows, pigs, horses and the like) and laboratory animals (e g , rats, mice, guinea pigs and the like)
  • Subjects "m need of treatment” with a compound of the present disclosure, or subjects “in need of NHE inhibition” include subjects with diseases and/or conditions that can be treated with substantially impermeable or substantially systemically non-bioavailable NHE-mhibiting compounds, with or without a fluid- absorbmg polymer, to achieve a beneficial therapeutic and/or prophylactic result
  • a beneficial outcome includes a decrease in the seventy of symptoms or delay m the onset of symptoms, increased longevity and/or more rapid or more complete resolution of the disease or condition
  • a subject in need of treatment is suffenng from a gastrointestinal tract disorder
  • the patient is suffenng from a disorder selected from the group consisting of a gastrointestinal motility disorder, irntable bowel syndrome, chronic constipation, chrome idiopathic constipation, chronic constipation occurnng in cystic fibrosis patients, chronic constipation occurnng in chronic kidney disease patients, calcium-induced constipation in osteoporotic patients, opioid-mduced constipation
  • the compounds described herein can be used alone or in combination with other agents
  • the compounds can be administered together with a diuretic (i e , High Ceiling Loop Diuretics, Benzothiadiazide Diuretics, Potassium Sparing Diuretics, Osmotic Diuretics), cardiac glycoside, ACE inhibitor, angiotensin-2 receptor antagonist, calcium channel blocker, beta blocker, alpha blocker, central alpha agonist, vasodilator, blood thinner, anti-platelet agent, hpid-lowe ⁇ ng agent, peroxisome prohferator-activated receptor (PPAR) gamma agonist agent or compound or with a fluid-absorbing polymer as more fully desc ⁇ bed below
  • the agent can be covalently attached to a compound desc ⁇ bed herein or it can be a separate agent that is administered together with or sequentially with a compound descnbed herein in a combination therapy
  • Combination therapy can be achieved by administering two or more agents, e g , a substantially non-permeable or substantially systemically non- bioavailable NHE-inhibiting compound desc ⁇ bed herein and a diuretic, cardiac glycoside, ACE inhibitor, angiotensm-2 receptor antagonist, calcium channel blocker, beta blocker, alpha blocker, central alpha agonist, vasodilator, blood thinner, antiplatelet agent or compound, each of which is formulated and administered separately, or by admmistenng two or more agents in a single formulation
  • two agents can be formulated together and administered in conjunction with a separate formulation containing a third agent While the two or more agents in the combination therapy can be administered simultaneously, they need not be
  • administration of a first agent (or combination of agents) can precede administration of a second agent (or combination of agents) by minutes, hours, days, or weeks
  • the two or more agents can be administered withm minutes of each other or within 1, 2, 3, 6, 9, 12, 15, 18,
  • Combination therapy can also include two or more administrations of one or more of the agents used in the combination
  • agent X and agent Y are used m a combination, one could administer them sequentially in any combination one or more times, e g , in the order X-Y-X, X-X-Y, Y-X-Y, Y Y-X, X-X-Y-Y, etc
  • the compounds desc ⁇ bed herein can be used in combination therapy with a diuretic
  • the useful analgesic agents are, for example High Ceiling Loop Diuretics [Furosemide (Lasix), Ethacrynic Acid (Edecnn) ,Bumetamde (Bumex)], Benzothiadiazide Diuretics [Hydrochlorothiazide (Hydrodm ⁇ l), Chlorothiazide (Diu ⁇ l), Clorthahdone (Hygroton), Benzthiazide (Aguapres), Bendroflumethiazi
  • Cardiac glycosides cardenohdes or other digitalis preparations can be administered with the compounds of the disclosure in co therapy
  • useful cardiac glycosides are, for example Digitoxm (Crystodigin), Digoxin (Lanoxin) or Deslanoside (Cedilamd-D) Cardiac glycosides in the va ⁇ ous classes are desc ⁇ bed in the literature
  • Angiotensin Converting Enzyme Inhibitors can be administered with the compounds of the disclosure in co-therapy
  • useful ACE inhibitors are, for example Captop ⁇ l (Capoten), Enalap ⁇ l (Vasotec), Lisinop ⁇ l (P ⁇ nivil) ACE inhibitors in the va ⁇ ous classes are desc ⁇ bed in the literature
  • Angiotensin-2 Receptor Antagonists also referred to as ATi -antagonists or angiotensin receptor blockers, or ARB 's
  • the useful Angiotensm-2 Receptor Antagonists are, for example Candesartan (Atacand), Eprosartan (Teveten), Irbesartan (Avapro), Losartan (Cozaar), Telmisartan (Micardis), Valsartan (Diovan)
  • Angiotensm-2 Receptor Antagonists in the various classes are desc ⁇ bed
  • PPAR gamma agonists such as thiazohdinediones (also called ghtazones) can be administered with the compounds of the disclosure in co-therapy
  • useful PPAR agonists are, for example rosightazone (Avandia), pioglitazone (Actos) and nvoghtazone
  • Aldosterone antagonists can be administered with the compounds of the disclosure m co-therapy
  • useful Aldosterone antagonists are, for example eplerenone, spironolactone, and canrenone
  • Alpha blockers can be administered with the compounds of the disclosure in co-therapy Among the useful Alpha blockers are, for example Doxazosin mesylate (Cardura), Prazosin hydrochlo ⁇ de (Mimpress) Prazosin and polythiazide (Mimzide), Terazosin hydrochloride (Hytrin) Alpha blockers in the various classes are descnbed in the literature
  • Central alpha agonists can be administered with the compounds of the disclosure in co-therapy
  • the useful Central alpha agonists are, for example Clomdine hydrochlo ⁇ de (Catapres), Clomdine hydrochloride and chlorthalidone (Clorpres, Combipres), Guanabenz Acetate (Wytensin), Guanfacine hydrochlo ⁇ de (Tenex), Methyldopa (Aldomet), Methyldopa and chlorothiazide (Aldochlor), Methyldopa and hydrochlorothiazide (Aldo ⁇ l)
  • Central alpha agonists in the various classes are descnbed in the literature
  • Vasodilators can be administered with the compounds of the disclosure in co-therapy
  • the useful vasodilators are, for example Isosorbide dimtrate
  • Blood thinners can be administered with the compounds of the disclosure in co-therapy Among the useful blood thinners are, for example Warfann (Coumadin) and Hepann Blood thinners in the vanous classes are descnbed in the literature
  • Anti-platelet agents can be administered with the compounds of the disclosure in co-therapy
  • useful anti-platelet agents are, for example Cyclooxygenase inhibitors (Aspi ⁇ n), Adenosine diphosphate (ADP) receptor inhibitors [Clopidogrel (Plavix), Ticlopidine (Ticlid)], Phosphodiesterase inhibitors [Cilostazol (Pletal)], Glycoprotein IIB/IIIA inhibitors [Abciximab (ReoPro), Eptifibatide (Integnlin), Tirofiban (Aggrastat), Defibrotide], Adenosine reuptake inhibitors [Dipyndamole (Persantine)]
  • Anti-platelet agents m the vanous classes are descnbed m the literature
  • Lipid-lowenng agents can be administered with the compounds of the disclosure in co-therapy
  • useful lipid-lowenng agents are, for example Statins (HMG CoA reductase inhibitors), [Atorvastatm (Lipitor), Fluvastatm (Lescol), Lovastatm (Mevacor, Altoprev), Pravastatin (Pravachol), Rosuvastatm Calcium (Crestor), Simvastatin (Zocor)], Selective cholesterol absorption inhibitors [ezetimibe (Zetia)], Resms (bile acid sequestrant or bile acid-binding drugs) [Cholestyramine (Questran, Questran Light, Prevalite, Locholest, Locholest Light), Colestipol (Colestid), Colesevelam HcI (WelChol)], Fibrates (Fib ⁇ c acid derivatives) [Gemfibrozil (Lopid), Fenofibrate (Antara, Lofibra, T ⁇ cor
  • the compounds of the disclosure can be used in combination with peptides or peptide analogs that activate the Guanylate Cyclase-receptor in the intestine and results in elevation of the intracellular second messenger, or cyclic guanosme monophosphate (cGMP), with increased chloride and bicarbonate secretion into the intestinal lumen and concomitant fluid secretion
  • cGMP cyclic guanosme monophosphate
  • Example of such peptides are Linaclotide (MD-1100 Acetate), endogenous hormones guanylm and uroguanylm and ente ⁇ c bacte ⁇ al peptides of the heat stable enterotoxm family (ST peptides) and those desc ⁇ bed in US 5140102, US 5489670, US 5969097, WO 2006/001931A2, WO 2008/002971A2, WO 2008/106429A2, US 2008/0227685A1 and US 7041786, the entire contents of which are
  • the compounds of the disclosure can be used in combination with type 2 chlo ⁇ de channel agonists, such as Amitiza (Lubiprostone) and other related compounds desc ⁇ bed in US 6414016, the entire contents of whch are incorporated herein by reference for all relevant and consistent purposes
  • the compounds of the disclosure can be used m combination with P2Y2 receptor agonists, such as those desc ⁇ bed in EP 1196396B1 and US 6624150, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes
  • Other agents include natnuretic peptides such as nesintide, a recombinant form of brain-nat ⁇ uretic peptide (BNP) and an at ⁇ al-nat ⁇ uretic peptide (ANP)
  • Vasopressin receptor antagonists such as tolvaptan and conivaptan may be coadministered as well as phosphate binders such as renagel, renleva, phoslo and fosrenol
  • Other agents include phosphate transport inhibitors (as desc ⁇ bed in U S Pat Nos 4,806,532, 6,355,823, 6,787,528, 7,119,120, 7,109,184, U S Pat Pub No 2007/021509, 2006/0280719
  • the compounds described herein can be used alone or in combination with other agents
  • the compounds can be administered together with an analgesic peptide or compound
  • the analgesic peptide or compound can be covalently attached to a compound desc ⁇ bed herein or it can be a separate agent that is administered together with or sequentially with a compound descnbed herein in a combination therapy
  • Combination therapy can be achieved by administering two or more agents, e g , a substantially non-permeable or substantially non-bioavailable NHE- inhibiting compound desc ⁇ bed herein and an analgesic peptide or compound, each of which is formulated and administered separately, or by administering two or more agents in a single formulation
  • two agents can be formulated together and administered in conjunction with a separate formulation containing a third agent
  • the two or more agents in the combination therapy can be administered simultaneously, they need not be
  • administration of a first agent (or combination of agents) can precede administration of a second agent (or combination of agents) by minutes, hours, days, or weeks
  • the two or more agents can be administered within minutes of each other or within 1, 2, 3, 6, 9, 12, 15, 18, or 24 hours of each other or within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14 days of each other or within 2, 3, 4, 5, 6, 7, 8, 9, or weeks of each other In some cases even longer intervals are possible While in many cases it is desirable that the
  • Combination therapy can also include two or more administrations of one or more of the agents used in the combination
  • agent X and agent Y are used in a combination, one could administer them sequentially in any combination one or more times, e g , in the order X-Y-X, X-X-Y, Y-X-Y, Y-Y-X, X-X-Y-Y, etc
  • the compounds desc ⁇ bed herein can be used in combination therapy with an analgesic agent, e g , an analgesic compound or an analgesic peptide
  • an analgesic agent e g , an analgesic compound or an analgesic peptide
  • the 5 analgesic agent can optionally be covalently attached to a compound desc ⁇ bed herein
  • the useful analgesic agents are, for example Ca channel blockers, 5HT3 agonists (e g , MCK-733), 5HT4 agonists (e g , tegaserod, prucalop ⁇ de), and 5HTl receptor antagonists, opioid receptor agonists (loperamide, fedotozine, and fentanyl), NKl receptor antagonists, CCK receptor agonists (e g , loxiglumide), NKl receptor
  • 5HT3 agonists e g , MCK-733
  • 5HT4 agonists e g ,
  • Opioid receptor antagonists and agonists can be administered with the compounds of the disclosure in co-therapy or linked to the compound of the disclosure,
  • t ⁇ mebutme which is thought to bind to mu/delta/kappa opioid receptors and activate release of motihn and modulate the release of gast ⁇ n, vasoactive intestinal peptide, gastrin and glucagons K opioid receptor agonists such as fedotozine, ketocyclazocme, and compounds desc ⁇ bed in US
  • ⁇ -opioid receptor agonists such as morphine, diphenyloxylate, frakefamide (H-Tyr-D-Ala-Phe(F)-Phe-NH 2 , disclosed in WO 01/019849 Al, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes) and loperamide can be used Tyr-Arg (kyotorphm) is a dipeptide that acts by stimulating the release of met-enkephahns to elicit an analgesic effect (J Biol Chem 262 8165, 1987) Kyotorphm can be used with or linked to the compounds of the disclosure
  • CCK receptor agonists such as caerulein from amphibians and other species are useful analgesic agents that can be used with or linked to the compounds of the disclosure
  • Peptide analogs of thymuhn can have analgesic activity and can be used with or linked to the compounds of the disclosure
  • CCK (CCKa or CCKb) receptor antagonists including loxiglumide and dexloxiglumide (the R-isomer of loxiglumide) (US 5,130,474 or WO 88/05774, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes) can have analgesic activity and can be used with or linked to the compounds of the disclosure
  • 5-HT4 agonists such as tegaserod/zelnorm and hrexapnde
  • 5-HT4 agonists such as tegaserod/zelnorm and hrexapnde
  • tegaserod/zelnorm Such agonists are described in EP 1321142 Al, WO 03/053432A1, EP 505322 Al, EP 505322 Bl, EP 507672 Al, EP 507672 Bl, U S Pat No 5,510,353 and U S Pat No 5,273,983, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes
  • Calcium channel blockers such as ziconotide and related compounds desc ⁇ bed in, for example, EP 625162Bl, U S Pat No 5,364,842, U S Pat No 5,587,454, U S Pat No 5,824,645, U S Pat No 5,859,186, U S Pat No 5,994,305, U S Pat No 6,087,091, U S Pat No 6,136,786, WO 93/13128 Al, EP 1336409 Al, EP 835126 Al, EP 835126 Bl, U S Pat No 5,795,864, U S Pat No 5,891,849, U S Pat No 6,054,429, WO 97/01351 Al, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes, can be used with or linked to the compounds of the disclosure
  • NKl receptor antagonists such as aprepitant (Merck & Co Inc), vofopitant, ezlopitant (Pfizer, Inc ), R-673 (Hoffmann-La Roche Ltd), SR-14033 and related compounds desc ⁇ bed in, for example, EP 873753 Al, U S 20010006972 Al, U S 20030109417 Al, WO 01/52844 Al, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes, can be used with or linked to the compounds of the disclosure
  • NK-2 receptor antagonists such as nepadutant (Mena ⁇ m Ricerche SpA), saredutant (Sanofi-Synthelabo), SR-144190 (Sanofi-Synthelabo) and UK-290795 (Pfizer Inc) can be used with or linked to the compounds of the disclosure
  • NK3 receptor antagonists such as osanetant (Sanofi-Synthelabo), talnetant and related compounds descnbed in, for example, WO 02/094187 A2, EP 876347 Al, WO 97/21680 Al, U S Pat No 6,277,862, WO 98/11090, WO 95/28418, WO 97/19927, and Boden et al (J Med Chem 39 1664-75, 1996) , the entire contents of which are incorporated herein by reference for all relevant and consistent purposes, can be used with or linked to the compounds of the disclosure
  • Norepinephrme-serotonin reuptake inhibitors such as milnacipran and related compounds described in WO 03/077897 Al, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes, can be used with or linked to the compounds of the disclosure
  • Vamlloid receptor antagonists such as arvaml and related compounds desc ⁇ bed m WO 01/64212 Al, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes, can be used with or linked to the compounds of the disclosure
  • the compounds can be used in combination therapy with a phosphodiesterase inhibitor (examples of such inhibitors can be found in U S Pat No 6,333,354, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes)
  • the compounds can be used alone or in combination therapy to treat disorders associated with chloride or bicarbonate secretion that may lead to constipation, e g , Cystic Fibrosis
  • the compounds can also or alternatively be used alone or in combination therapy to treat calcium-mduced constipation effects
  • Constipation is commonly found in the geriatric population, particularly patients with osteoporosis who have to take calcium supplements
  • Calcium supplements have shown to be beneficial in ostoporotic patients to restore bone density but compliance is poor because of constipation effects associated therewith
  • Opioid use is mainly directed to pam relief, with a notable side-effect being GI disorder, e g constipation
  • GI disorder e g constipation
  • opioid receptors which are found principally in the central nervous system and the gastrointestinal tract
  • the receptors in these two organ systems mediate both the beneficial effects, and the undesirable side effects (e g decrease of gut motility and ensuing constipation)
  • Opioids suitable for use typically belong to one of the following exemplary classes natural opiates, alkaloids contained in the resin of the opium poppy including morphine, codeine and thebaine, semi-synthetic opiates, created from the natural opioids, such as hydromorphone, hydrocodone, oxycodone, oxymorphone, desomorphme, diacetylmorphine (Heroin), mcomorphme, dipropanoylmorphine, benzylmorphine and ethylmorphine, fully synthetic opioids, such as fent
  • the compound of the disclosure can be used alone or in combination therapy to alleviate GI disorders encountered with patients with renal failure (stage 3- 5) Constipation is the second most reported symptom in that category of patients (Murtagh et al , 2006, Murtagh et al , 2007a, Murtagh et al , 2007b) Without being held by theory, it is believed that kidney failure is accompanied by a stimulation of intestinal Na re-absorption (Hatch and Freel, 2008) A total or partial inhibition of such transport by administration of the compounds of the disclosure can have a therapeutic benefit to improve GI transit and relieve abdominal pain
  • the compounds of the disclosure can be used in combination with Angiotensin-modulating agents Angiotensin Converting Enzyme (ACE) inhibitors (e g captop ⁇ l, enalop ⁇ l, 5 hsmop ⁇ l, ramip ⁇ l) and Angiotensin II receptor antagonist therapy (also referred to as ATi-antagonists or angiotensin receptor blockers,
  • Phoslo, Fosrenol phosphate transport inhibitor such as those desc ⁇ bed in US 4806532, US 6355823, US 6787528, WO 2001/005398, WO 2001/087294, WO 2001/082924, WO 2002/028353, WO 2003/048134, WO 2003/057225, US 7119120, EP 1465638, US Appl 2007/021509, WO 2003/080630, US 7109184, US Appl 2006/0280719 , EP 1485391, WO 2004/085448, WO 2004/085382, US Appl 2006/0217426, JP
  • the compounds of the disclosure can be used in combination with peptides or peptide analogs that activate the Guanylate Cyclase-receptor in the intestine and results in elevation of the intracellular second messenger, or cyclic guanosine
  • cGMP monophosphate
  • ST peptides the heat stable enterotoxm family
  • the compounds of the disclosure can be used in combination with type-2 chlo ⁇ de channel agonists, such as Amitiza (Lubiprostone) and other related compounds described in US 6414016, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes
  • P2Y2 receptor agonists such as those desc ⁇ bed in EP 1196396B1 and US 6624150, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes
  • the compounds of the disclosure can be used in combination with laxative agents such as bulk-producing agents, e g psyllium husk (Metamucil), methylcellulose (Citrucel), polycarbophil, dietary fiber, apples, stool softeners/surfactant such as docusate (Colace, Diocto), hydrating agents (osmotics), such as dibasic sodium phosphate, magnesium citrate, magnesium hydroxide (Milk of magnesia), magnesium sulfate (which is Epsom salt), monobasic sodium phosphate, sodium biphosphate, hyperosmotic agents glycenn suppositones, sorbitol, lactulose, and polyethylene glycol (PEG)
  • laxative agents such as bulk-producing agents, e g psyllium husk (Metamucil), methylcellulose (Citrucel), polycarbophil, dietary fiber, apples, stool softeners/surfactant such as docusate (Colace,
  • the compounds of the disclosure accelerate gastrointestinal transit, and more specifically in the colon, without substantially affecting the residence time in the stomach, i e with no significant effect on the gast ⁇ c emptying time
  • the compounds of the invention restore colonic transit without the side-effects associated with delayed gastric emptying time, such as nausea
  • the GI and colonic transit are measured in patients using methods reported in, for example Burton DD, Camille ⁇ M, Mullan BP, et al , J Nucl Med , 1997,38 1807- 1810, Cremonini F, Mullan BP, Camille ⁇ M, et al , Aliment Pharmacol Ther , 2002,16 1781-1790, Camille ⁇ M, Zinsmeister AR, Gastroenterology, 1992,103 36-42, Bouras EP, Camille ⁇ M, Burton DD, et al , Gastroenterology, 2001,120 354-360, Coulie B, Szarka LA, Camille ⁇ M, et al , Gastroenterology, 2000
  • the NHE-inhibiting compounds desc ⁇ bed therein may be administered to patients in need thereof in combination with a fluid-absorbing polymer ("FAP")
  • the intestinal fluid-absorbing polymers useful for administration in accordance with embodiments of the present disclosure may be administered orally in combination with non-absorbable NHE-inhibitors (e g , a NHE-3 inhibitor) to absorb the intestinal fluid resulting from the action of the sodium transport inhibitors
  • non-absorbable NHE-inhibitors e g , a NHE-3 inhibitor
  • Such polymers swell in the colon and bind fluid to impart a consistency to stools that is acceptable for patients
  • the fluid-absorbing polymers desc ⁇ bed herein may be selected from polymers with laxative properties, also referred to as bulking agents (i e , polymers that retain some of the intestinal fluid m the stools and impart a higher degree of hydration in the stools and facilitate transit)
  • the flmd-absorbmg polymers may also be optionally
  • the ability of the polymer to maintain a certain consistency in stools with a high content of fluid can be characterized by its "water holding power " Wenzl et al (in Determinants of decreased fecal consistency in patients with diarrhea, Gastroenterology, v 108, no 6, p 1729-1738 (1995)) studied the determinants that control the consistency of stools of patients with diarrhea and found that they were narrowly correlated with the water holding power of the feces
  • the water holding power is determined as the water content of given stools to achieve a certain level of consistency (corresponding to "formed stool" consistency) after the reconstituted fecal matter has been cent ⁇ fuged at a certain g number
  • the water holding power of the feces is increased by ingestion of certain polymers with a given fluid absorbing profile More specifically, it has been found that the water-holding power of said polymers is correlated with their fluid absorbancy under load (AUL), even more specifically the AUL of said polymers is greater than
  • the FAP utilized in the treatment method of the present disclosure preferably has a AUL of at least about 1O g, about 15 g, about 2O g, aboug 25 g or more of isotonic fluid/g of polymer under a static pressure of about 5 kPa, and preferably about 10 kPA, and may have a fluid absorbency of about 20 g, about 25 g or more, as determined using means generally known in the art Additionally or alternatively, the FAP may impart a minimum consistency to fecal matter and, in some embodiments, a consistency graded as "soft" in the scale described in the test method below, when fecal non water-soluble solid fraction is from 10% to 20%, and the polymer concentration is from 1% to 5% of the weight of stool The determination of the fecal non water-soluble solid fraction of stools is described in Wenz et al
  • the polymer may be uncharged or may have a low charge density (e g , 1-2 meq/gr) Alternatively or
  • the FAP is a "superabsorbent" polymer (i e , a lightly crosslinked, partially neutralized polyelectrolyte hydrogel similar to those used in baby diapers, feminine hygiene products, agriculture additives, etc )
  • Superabsorbent polymers may be made of a lightly crosslinked polyacrylate hydrogel
  • the swelling of the polymer is dnven essentially by two effects (i) the hydration of the polymer backbone and entropy of mixing and (ii) the osmotic pressure arising from the counter-ions (e g , Na ions) within the gel
  • the gel swelling ratio at equilibrium is controlled by the elastic resistance inherent to the polymer network and by the chemical potential of the bathing fluid, i e , the gel will de-swell at higher salt concentration because the background electrolyte will reduce the apparent charge density on the polymer and will reduce the difference of free ion concentrations inside and outside the gel that d ⁇ ves osmotic pressure
  • the swelling ratio SR
  • the degree of crosslinking can vary greatly depending upon the specific polymer material, however, m most applications the subject superabsorbent polymers are only lightly crosslinked, that is, the degree of crosslinking is such that the polymer can still absorb over 10 times its weight in physiological saline (i e , 0 9% saline)
  • such polymers typically include less than about 02 mole % crosslinking agent
  • the FAP's utilized for treatment are Calcium Carbophil (Registry Number 9003-97-8, also referred as Carbopol EX-83), and Carpopol 934P
  • the fluid-absorbmg polymer is prepared by high internal phase emulsion ("HIPE") processes
  • HIPE high internal phase emulsion
  • the HIPE process leads to polymeric foam slabs with a very large porous fraction of interconnected large voids (about 100 microns) (i e , open-cell structures)
  • This technique produces flexible and collapsible foam mate ⁇ als with exceptional suction pressure and fluid absorbency (see U S Patent Nos 5,650,222, 5,763,499 and 6,107,356, which are incorporated herein for all relevant and consistent purposes)
  • the polymer is hydrophobic and, therefore, the surface should be modified so as to be wetted by the aqueous fluid This is accomplished by post-treating the foam mate ⁇ al by a surfactant in order to reduce the interfacial tension
  • fluid-absorbmg gels are prepared by aqueous free radical polymerization of acrylamide or a de ⁇ vative thereof, a crosslinker (e g , methylene-bis-acrylamide) and a free radical initiator redox system in water
  • a crosslinker e g , methylene-bis-acrylamide
  • a free radical initiator redox system in water
  • the mate ⁇ al is obtained as a slab
  • the swelling ratio of crosslmked polyacrylamide at low crosslinking density e g , 2%-4% expressed as weight % of methylene-bis-acrylamide
  • the swelling properties of these polymers have been extensively studied and are essentially the same of those of crosslmked polyacrylic acids at high salt concentration Under those conditions, the osmotic pressure is null due to the presence of counter-ions and the swelling is controlled by the free energy of mixing and the network elastic energy Stated differently, a crosslmked polyacrylamide gel of same crosslink density as a neutralized
  • the FAP utilized for treatment in combination with a NHE-mhibitor is a superporous gel that may delay the emptying of the stomach for the treatment of obesity (J Chen, Journal of Controlled Release, 65, pp 73-82 (2000), or to deliver proteins
  • Polyacrylate-based SAP's with a macroporous structure may also be used
  • Macroporous SAP and superporous gels differ in that the porous structure remains almost intact in the dry state for superporous gels, but disappears upon drying for macroporous SAP's
  • the method of preparation is different although both methods use a foaming agent (e g , carbonate salt that generates CO 2 bubbles du ⁇ ng polymerization)
  • Typical swelling ratios, SR, of superporous matenals are around 10 Superporous gels keep a large internal pore volume in the dry state
  • Macroporous hydrogels may also be formed using a method whereby polymer phase separation in induced by a non-solvent
  • the polymer may be polyNIPAM and the non-solvent utilized may be glucose (see, e g , Z Zhang, J Org Chem , 69, 23 (2004)) or NaCl (see, e g , Cheng et al , Journal of Biomedical Matenals Research - Part A, VoI 67, Issue 1, 1 October 2003, Pages 96-103)
  • the phase separation induced by the presence of NaCl leads to an increase in swelling ratio
  • These matenals are preferred if the swelling ratio of the material, SR, is maintained in salt isotonic solution and if the gels do not collapse under load
  • the temperature of "service” should be shifted beyond body temperature, e g by diluting NIPAM in the polymer with monomer devoid of transition temperature phenomenon
  • the fluid-absorbing polymer may be selected from certain naturally-occurring polymers such as those containing carbohydrate moieties In a preferred embodiment, such carbohydrate-containing hydrogels are non- digestible, have a low fraction of soluble mate ⁇ al and a high fraction of gel-formmg materials
  • the fluid-absorbmg polymer is selected from xanthan, guar, wellan, hemicelluloses, alkyl-cellulose, hydro-alkyl-cellulose, carboxy-alkyl- cellulose, carrageenan, dextran, hyaluronic acid and agarose
  • the gel forming polymer is psyllium Psyllium (or "lspaghula") is the common name used for several members of the plant genus Plantago whose seeds are used commercially for the production of mucilage
  • the fluid-absorbing polymer is in the gel-formmg fraction of psyllium, i e ,
  • the psyllium-contaimng dosage form includes physically discrete unit suitable as a unitary dosage for human subjects and other mammals, each containing a predetermined quantity of active mate ⁇ al (e g the gel-forming polysaccharide) calculated to produce the desired therapeutic effect
  • Solid oral dosage forms that are suitable for the present compositions include tablets, pills, capsules, lozenges, chewable tablets, troches, cachets, pellets, wafer and the like
  • the FAP is a polysaccharide particle wherein the polysaccha ⁇ de component includes xylose and arabmose The ratio of the xylose to the arabinose may be at least about 3 1 by weight, as described in U S Pat Nos 6,287,609, 7,026,303 and 7,014,862, each of which is incorporated herein for all relevant and consistent purposes
  • the fluid-absorbing polymers desc ⁇ bed herein may be used in combination with the NHE-mhibiting compounds or a pharmaceutical composition containing the compound
  • the NHE inhibitor and the FAP may also be administered with other agents including those desc ⁇ bed under the heading "Combination Therapies" without departing from the scope of the present disclosure
  • the NHE inhibitor may be administered alone without use of a fluid-absorbing polymer to resolve symptoms without eliciting significant diarrhea or fecal fluid secretion that would require the co-admimstration of a fluid-absorbing polymer
  • the fluid-absorbing polymers desc ⁇ bed herein may be selected so as to not induce any substantial interaction with the NHE-inhibitmg compounds or a pharmaceutical composition containing the compound
  • "no substantial interaction” generally means that the co-admimstration of the FAP polymer would not substantially alter (i e , neither substantially decrease nor substantially increase) the pharmacological property of the NHE-inhibiting compounds administered alone
  • FAPs containing negatively charged functionality such as carboxylates, sulfonates, and the like, may potentially interact ionically with positively charged NHE inhibitors, preventing the inhibitor from reaching its pharmacological target
  • the shape and arrangement of functionality in a FAP could act as a molecular recognition element, and sequestor NHE inhibitors via "host- guest” interactions via the recognition of specific hydrogen bonds and/or hydrophobic regions of a given inhibitor Accordingly, in vanous embodiments of the present disclosure, the FAP polymer may be selected, for co-admimst
  • “pharmaceutically effective amount” of a compound disclosed herein is a quantity that results m a beneficial clinical outcome of the condition being treated with the compound compared with the absence of treatment
  • the amount of the compound or compounds administered will depend on the degree, seventy, and type of the disease or condition, the amount of therapy desired, and the release characte ⁇ stics of the pharmaceutical formulation It will also depend on the subject's health, size, weight, age, sex and tolerance to drugs Typically, the compound is administered for a sufficient pe ⁇ od of time to achieve the desired therapeutic effect
  • the NHE-inhibitor and FAP may be administered together or in a "dual-regimen" wherein the two therapeutics are dosed and administered separately
  • the typical dosage administered to the subject in need of the NHE inhibitor is typically from about 5 mg per day and about 5000 mg per day and, in other embodiments, from about 50 mg per day and about 1000 mg per day
  • Such dosages may induce fecal excretion of sodium (and its accompanying anions), from about 10 mmol up to about 250 mmol per day, from about 20 mmol to about 70 mmol per day or even from about 30 mmol to about 60 mmol per day
  • the typical dose of the fluid-absorbing polymer is a function of the extent of fecal secretion induced by the non-absorbable NHE inhibitor Typically the dose is adjusted according to the frequency of bowel movements and consistency of the stools More specifically the dose is adjusted so as to avoid liquid stools and maintain stool consistency as "soft" or semi-formed, or formed
  • typical dosage ranges of the fluid- absorbing polymer to be administered in combination with the NHE inhibitor are from about 2 g to about 50 g per day, from about 5 g to about 25 g per day or even from about 10 g to about 20 g per day
  • the daily uptake may be from about 2 g to about 50 g per day, from about 5 g to about 25 g per day, or from about 10 g to about 20 g per day, with a weight ratio of NHE inhibitor to fluid- absorbing polymer being from 5 about 1 1000 to 1 10
  • a typical dosage of the substantially impermeable or substantially systemically non-bioavailable, NHE-mhibiting compound when used alone without a FAP may be between about 0 2 mg per day and about 2 g per day, or between about 1 mg and about 1 g per day, or between about 5 mg and about 500 mg, or between about
  • the frequency of administration of therapeutics desc ⁇ bed herein may vary from once-a-day (QD) to twice-a-day (BID) or fh ⁇ ce-a-day (TID), etc , the precise frequency of administration varying with, for example, the patient's condition, the
  • the NHE-mhibitor could be taken once-a-day while the fluid-absorbmg polymer could be taken at each meal (TID)
  • the substantially impermeable or substantially systemically non- 20 bioavailable, NHE-inhibiting compounds of the present disclosure with or without the fluid-absorbing polymers desc ⁇ bed herein may be administered by any suitable route
  • the compound is preferably administrated orally (e g , dietary) in capsules, suspensions, tablets, pills, dragees, liquids, gels, syrups, slurries, and the like
  • Methods for encapsulating compositions are 25 known in the art (Baker, et al , "Controlled Release of Biological Active Agents", John Wiley and Sons, 1986)
  • the compounds can be administered to the subject in conjunction with an acceptable pharmaceutical earner as part of a pharmaceutical composition
  • the formulation of the pharmaceutical composition will vary according to the route of administration selected Suitable pharmaceutical earners may contain 3 0 inert ingredients which do not interact with the compound
  • the earners are biocompatible, i e , non-toxic, non-mflammatory, non-immun
  • Examples of pharmaceutically acceptable earners include, for example, saline, commercially available inert gels, or liquids supplemented with albumin, methyl cellulose or a collagen matrix Standard pharmaceutical formulation techniques can be employed, such as those described in 5 Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa
  • compositions for oral use can be obtained by combining a compound of the present disclosure with a solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • suitable excipients are, in
  • fillers such as sugars, including lactose, sucrose, manmtol, or sorbitol, cellulose preparations such as, for example, maize starch, wheat starch, ⁇ ce starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP)
  • PVP polyvinylpyrrolidone
  • disintegrating agents can be added, such as cross-linked polyvinyl pyrrolidone, agar, or
  • Dragee cores are provided with suitable coatings
  • suitable coatings for this purpose, concentrated sugar solutions can be used, which can optionally contain gum arable, talc, polyvinyl pyrrohdone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures Dyestuffs or
  • 2Q pigments can be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses
  • compositions which can be used orally include push-fit capsules made of a suitable mate ⁇ al, such as gelatin, as well as soft, sealed capsules made of a suitable mate ⁇ al, for example, gelatin, and a plasticizer, such as glycerol or
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lub ⁇ cants such as talc or magnesium stearate and, optionally, stabilizers
  • filler such as lactose, binders such as starches, and/or lub ⁇ cants such as talc or magnesium stearate and, optionally, stabilizers
  • the active compounds can be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols
  • stabilizers can be added All formulations for
  • NHE proteins show considerable diversity in their patterns of tissue expression, membrane localization and functional roles (See, e g , The sodium- hydrogen exchanger - From molecule To Its Role In Disease, Karmazyn, M , Avkiran, M , and Fhegel, L , eds , Kluwer Academics (2003) )
  • NHE-I through -9 nine distinct NHE genes (NHE-I through -9) have been descnbed Of these nine, five (NHE-I through -5) are principally active at the plasma membrane, whereas NHE-6, -7 and -9 reside predominantly within intracellular compartments
  • NHE-I is ubiquitously expressed and is chiefly responsible for restoration of steady state intracellular pH following cytosolic acidification and for maintenance of cell volume
  • NHE-I is crucial for organ function and survival (e g NHE-I -null mice exhibit locomotor abnormalities, epileptic- hke seizures and considerable mortality before weaning)
  • NHE-2 through -4 are predominantly expressed on the apical side of epitheha of the kidney and the gastrointestinal tract Several lines of evidence show that NHE-3 is the major cont ⁇ butor of renal bulk Na+ and fluid re-absorption by the proximal tubule The associated secretion of H+ by NHE-3 into the lumen of renal tubules is also essential for about 2/3 of renal HCO3 re-absorption Complete disruption of NHE-3 function in mice causes a sharp reduction in HCO3 , Na+ and fluid re-absorption in the kidney, which is consistently associated with hypovolemia and acidosis
  • the novel compounds of the invention are intended to target the apical NHE antiporters (e g NHE-3, NHE-2 and NHE-8) without substantial permeability across the layer of gut epithelial cells, and/or without substantial activity towards NHEs that do not reside predominantly
  • the compounds of the invention are delivered to the small bowel with little or no interaction with the upper GI such as the gastric compartment and the duodenum
  • the compounds of the invention are designed so as to be released in an active form past the duodenum This can be accomplished by either a prodrug approach or by specific drug delivery systems
  • prodrug is to be understood to refer to a modified form of the compounds detailed herein that is inactive (or significantly less active) in the upper GI, but once administered is metabolised in vivo into an active metabolite after getting past, for example, the duodenum
  • the activity of the NHE inhibitor can be masked with a transient protecting group that is liberated after compound passage through the desired gast ⁇ c compartment
  • acylation or alkylation of the essential guanidinyl functionality of the NHE inhibitor would render it biochemically inactive, however, cleavage of these functional groups by intestinal amidases, esterases, phosphatases , and the like, as well enzymes present in the colonic flora, would liberate the active parent compound
  • Prodrugs can be designed to exploit the relative expression and localization of such phase I metabolic enzymes by carefully optimizing the structure of the prodrug for recognition by specific enzymes
  • the antiinflammatory agent sulfasal As an example, the antiinflammatory agent sulfasal
  • the NHE-inhibitor compounds of the invention are formulated in certain pharmaceutical compositions for oral administration that release the active in the targeted areas of the GI, i e , jejunum, ileum or colon, or preferably the distal ileum and colon, or even more preferably the colon
  • fluid-absorbing polymers that are administered m accordance with treatment methods of the present disclosure are formulated to provide acceptable/pleasant organoleptic properties such as mouthfeel, taste, and/or to avoid premature swelling/gelation in the mouth and in the esophagus and provoke choking or obstruction
  • the formulation may be designed in such a way so as to ensure the full hydration and swelling of the FAP in the GI tract and avoid the formation of lumps.
  • the oral dosages for the FAP may take various forms including, for example, powder, granulates, tablets, wafer, cookie and the like, and are most preferably delivered to the small bowel with little or no interaction with the upper GI such as the gastric compartment and the duodenum .
  • diethyl 2-aminoethylphosphonate Into a 500-mL round bottom S flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of diethyl 2-(l,3-dioxoisoindohn-2-yl)ethylphosphonate (5 g, 16 08 mmol, 1 00 equiv) in ethanol (200 mL) and hydrazine hydrate (8 g, 160 00 mmol, 9 95 equiv) The resulting solution was stirred for 12 h at room temperature The solids were filtered and the resulting mixture was concentrated under vacuum The residue was applied onto a0 silica gel column and eluted with dichloromethane/methanol (9 1) This resulted in 1 5 g (51%) of diethyl 2-aminoethylphosphonate as colorless oil
  • N-(4-(2-bromoacetyl)phenyl)acetamide Into a 100-mL 3- necked round-bottom flask, was placed a solution of N-(4-acetylphenyl)acetamide (1 g, 5 65 mmol, 1 00 equiv) in acetic acid (10 mL) This was followed by the addition of a solution of bromine (910 mg, 5 69 mmol, 1 01 equiv) in acetic acid (2 mL) dropwise with stirring at 5O 0 C The resulting solution was stirred for 1 5 h at 5O 0 C The reaction was then quenched by the addition of 100 mL of water/ice The solids were collected by filtration and d ⁇ ed under vacuum This resulted m 0 5 g (33%) of N-(4-(2- bromoacety])phenyl)acetamide as a white solid
  • Compound 53 (E)-4-(4-(4-(3-(diaminomethyleneamino)-2-methyl-3-oxoprop-l- enyl)-2,6-difluorophenoxy)phenylsulfonamido)benzylphosphonic acid.
  • Compound 53 was prepared from diethyl 4-aminobenzylphosphonate (intermediate 3.2) using the procedures described in Example 52 except the final product was purified by preparative HPLC.

Abstract

The present disclosure is directed to compounds and methods for the treatment of disorders associated with fluid retention or salt overload, such as heart failure (in particular, congestive heart failure), chronic kidney disease, end-stage renal disease, liver disease, and peroxisome proliferator-activated receptor (PPAR) gamma agonist-induced fluid retention. The present disclosure is also directed to compounds and methods for the treatment of hypertension. The present disclosure is also directed to compounds and methods for the treatment of gastrointestinal tract disorders, including the treatment or reduction of pain associated with gastrointestinal tract disorders. The methods generally comprise administering to a mammal in need thereof a pharmaceutically effective amount of a compound, or a pharmaceutical composition comprising such a compound, that is designed to be substantially active in the gastrointestinal (GI) tract to inhibit NHE-mediated antiport of sodium ions and hydrogen ions therein. More particularly, the method comprises administering to a mammal in need thereof a pharmaceutically effective amount of a compound, or a pharmaceutical composition comprising such a compound, that inhibits NHE-3, -2 and/or -8 mediated antiport of sodium and/or hydrogen ions in the GI tract and is designed to be substantially impermeable to the layer of epithelial cells, or more specifically the epithelium of the GI tract. As a result of the compound being substantially impermeable, it is not absorbed and is thus essentially systemically non-bioavailable, so as to limit the exposure of other internal organs (e.g., liver, heart, brain, etc.) thereto. The present disclosure is still further directed to a method wherein a mammal is administered such a compound with a fluid-absorbing polymer, such that the combination acts as described above and further provides the ability to sequester fluid and/or salt present in the GI tract.

Description

COMPOUNDS AND METHODS FOR INHIBITING NHE-MEDIATED ANTIPORT
IN THE TREATMENT OF DISORDERS ASSOCIATED WITH FLUID RETENTION
OR SALT OVERLOAD AND GASTROINTESTINAL TRACT DISORDERS
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit under 35 U S C §119(e) of U S
Provisional Patent Application No 61/141,853, filed December 31, 2008, U S
Provisional Patent Application No 61/169,509, filed Apπl 15, 2009, and U S
Provisional Patent Application No 61/237,842, filed August 28, 2009, which applications are incorporated herein by reference in their entireties
BACKGROUND
Field The present disclosure is directed to compounds that are substantially active in the gastrointestinal tract to inhibit NHE-mediated antiport of sodium ions and hydrogen ions, and the use of such compounds in the treatment of disorders associated with fluid retention or salt overload and in the treatment of gastrointestinal tract disorders, including the treatment or reduction of pain associated with a gastrointestinal tract disorder
Description of the Related Art
Disorders Associated with Fluid Retention and Salt Overload
According to the American Heart Association, more than 5 million Ameπcans have suffered from heart failure, and an estimated 550,000 cases of congestive heart failure (CHF) occur each year (Schocken, D D et al , Prevention of heart failure a scientific statement from the American Heart Association Councils on
Epidemiology and Prevention, Clinical Cardiology, Cardiovascular Nursing, and High
Blood Pressure Research, Quality of Care and Outcomes Research Interdisciplinary Working Group, and Functional Genomics and Translational Biology Interdisciplinary
Working Group Circulation, v 117, no 19, p 2544 2565 (2008)) The clinical syndrome of congestive heart failure occurs when cardiac dysfunction prevents adequate perfusion of peripheral tissues The most common form of heart failure leading to CHF is systolic heart failure, caused by contractile failure of the myocardium A mam cause of CHF is due to ischemic coronary artery disease, with or without infarction Long standing hypertension, particularly when it is poorly controlled, may lead to CHF
In patients with CHF, neurohumoral compensatory mechanisms (i e , the sympathetic nervous system and the renin-angiotensin system) are activated m an effort to maintain normal circulation The remn-angiotensm system is activated in response to decreased cardiac output, causing increased levels of plasma renin, angiotensin II, and aldosterone As blood volume increases in the heart, cardiac output increases proportionally, to a point where the heart is unable to dilate further In the failing heart, contractility is reduced, so the heart operates at higher volumes and higher filling pressures to maintain output Filling pressures may eventually increase to a level that causes transudation of fluid into the lungs and congestive symptoms (e g , edema, shortness of breath) All of these symptoms are related to fluid volume and salt retention, and this chronic fluid and salt overload further contnbute to disease progression
Compliance with the medication regimen and with dietary sodium restπctions is a cπtical component of self-management for patients with heart failure and may lengthen life, reduce hospitalizations and improve quality of life Physicians often recommend keeping salt intake below 2 3 g per day and no more than 2 g per day for people with heart failure Most people eat considerably more than this, so it is likely that a person with congestive heart failure will need to find ways to reduce dietary salt A number of drug therapies currently exist for patients suffeπng from
CHF For example, diuretics may be used or administered to relieve congestion by decreasing volume and, consequently, filling pressures to below those that cause pulmonary edema By counteracting the volume increase, diuretics reduce cardiac output, however, fatigue and dizziness may replace CHF symptoms Among the classes or types of diuretics currently being used is thiazides Thiazides inhibit NaCl transport in the kidney, thereby preventing reabsorption of Na in the cortical diluting segment at the ending portion of the loop of Henle and the proximal portion of the distal convoluted tubule However, these drugs are not effective when the glomerular filtration rate (GFR) is less than 30 ml/min Additionally, thiazides, as well as other diuretics, may cause hypokalemia Also among the classes or types of diuretics currently being used is loop diuretics (e g , furosemide) These are the most potent diuretics and are particularly effective in treating pulmonary edema Loop diuretics inhibit the NaKCl transport system, thus preventing reabsorption of Na in the loop of Henle
Patients that have persistent edema despite receiving high doses of diuretics may be or become diuretic-resistant Diuretic resistance may be caused by poor availability of the drug In patients with renal failure, which has a high occurrence in the CHF population, endogenous acids compete with loop diuretics such as furosemide for the organic acid secretory pathway in the tubular lumen of the nephron Higher doses, or continuous infusion, are therefore needed to achieve entrance of an adequate amount of drug into the nephron However, recent meta-analysis have raised awareness about the long-term πsk of chronic use of diuretics in the treatment of CHF For instance, in a recent study (Ahmed et al , Int J Cardiol 2008 Apπl 10, 125(2) 246 253) it was shown that chronic diuretic use was associated with significantly increased mortality and hospitalization in ambulatory older adults with heart failure receiving angiotensin converting enzyme inhibitor and diuretics
Angiotensm-converting enzyme ("ACE") inhibitors are an example of another drug therapy that may be used to treat congestive heart failure ACE inhibitors cause vasodilatation by blocking the renin-angiotensin-aldosterone system Abnormally low cardiac output may cause the renal system to respond by releasing renin, which then converts angiotensinogen into angiotensin I ACE converts angiotensin I into angiotensin II Angiotensin II stimulates the thirst centers in the hypothalamus and causes vasoconstπction, thus increasing blood pressure and venous return Angiotensin II also causes aldosterone to be released, causing reabsorption of Na and concomitant passive reabsorption of fluid, which in turn causes the blood volume to increase ACE inhibitors block this compensatory system and improve cardiac performance by decreasing systemic and pulmonary vascular resistance ACE inhibitors have shown survival benefit and conventionally have been a treatment of choice for CHF However, since ACE inhibitors lower aldosterone, the K-secreting hormone, one of the side-effects of their use is hyperkalemia In addition, ACE inhibitors have been show to lead to acute renal failure in certain categories of CHF patients (See, e g , C S Cruz et al , "Incidence and Predictors of Development of Acute Renal Failure Related to the Treatment of Congestive Heart Failure with ACE Inhibitors, Nephron CIm Pract , v 105, no 2, pp c77-c83 (2007))
Patients with end stage renal disease ("ESRD"), i e , stage 5 chronic kidney failure, must undergo hemodialysis three times per week The quasi-absence of renal function and ability to eliminate salt and fluid results m large fluctuations in body weight as fluid and salt build up in the body (sodium/volume overload) The fluid overload is characteπzed as mterdialytic weight gain High fluid overload is also worsened by heart dysfunction, specifically CHF Dialysis is used to remove uremic toxins and also adjust salt and fluid homeostasis However, symptomatic mtradialytic hypotension (SIH) may occur when patients are over-dialyzed SIH is exhibited in about 15% to 25% of the ESRD population (Davenport, A , C Cox, and R Thuraisingham, Blood pressure control and symptomatic mtradialytic hypotension in diabetic haemodialysis patients a cross-sectional survey, Nephron CIm Pract , v 109, no 2, p c65-c71 (2008)) Like in hypertensive and CHF patients, dietary restrictions of salt and fluid are highly recommended but poorly followed because of the poor palatability of low-salt food
The cause of pnmary or "essential" hypertension is elusive However, several observations point to the kidney as a pnmary factor The strongest data for excess salt intake and elevated blood pressure come from INTERSALT, a cross- sectional study of greater than 10,000 participants For individuals, a significant, positive, independent linear relation between 24-hour sodium excretion and systolic blood pressure was found Higher individual 24-hour uπnary sodium excretions were found to be associated with higher systolic/diastohc blood pressure on average, by 6- 3/3-0 mm Hg Pnmary hypertension is a typical example of a complex, multifactorial, and polygenic trait All these monogenic hypertensive syndromes are virtually confined to mutated genes involving gam of function of vanous components of the renin angiotensin-aldosterone system, resulting in excessive renal sodium retention In a broad sense, these syndromes are characteπzed by increased renal sodium reabsorption ansmg through either primary defects in sodium transport systems or stimulation of mmeralocortieoid receptor activity (Altun, B , and M Aπci, 2006, Salt and blood pressure time to challenge, Cardiology, v 105, no 1, p 9-16 (2006)) A much larger number of controlled studies have been performed on hypertensive subjects during the last three decades to determine whether sodium reduction will reduce established high blood pressure Meta-analyses of these studies have clearly shown a large decrease m blood pressure in hypertensive patients In end stage liver disease (ESLD), accumulation of fluid as ascites, edema or pleural effusion due to cirrhosis is common and results from a derangement in the extracellular fluid volume regulatory mechanisms Fluid retention is the most frequent complication of ESLD and occurs in about 50% of patients within 10 years of the diagnosis of cirrhosis This complication significantly impairs the quality of life of cirrhotic patients and is also associated with poor prognosis The one-year and five- year survival rate is 85% and 56%, respectively (Kashani et al , Fluid retention m cirrhosis pathophysiology and management, QJM, v 101, no 2, p 71-85 (2008)) The most acceptable theories postulate that the initial event m ascites formation in the cirrhotic patient is sinusoidal hypertension Portal hypertension due to an increase in sinusoidal pressure activates vasodilatory mechanisms In advanced stages of cirrhosis, arteriolar vasodilation causes underfilling of systemic arterial vascular space This event, through a decrease in effective blood volume, leads to a drop in arterial pressure Consequently, baroreceptor-mediated activation of renm-angiotensin aldosterone system, sympathetic nervous system and nonosmotic release of antidiuretic hormone occur to restore the normal blood homeostasis These events cause further retention of renal sodium and fluid Splanchnic vasodilation increases splanchnic lymph production, exceeding the lymph transportation system capacity, and leads to lymph leakage into the peπtoneal cavity Persistent renal sodium and fluid retention, alongside increased splanchnic vascular permeability in addition to lymph leakage into the peπtoneal cavity, play a major role in a sustained ascites formation Thiazolidmediones (TZD's), such as rosightazone, are peroxisome prohferator-activated receptor (PPAR) gamma agonist agents used for the treatment of type-2 diabetes and are widely prescnbed Unfortunately, fluid retention has emerged as the most common and serious side-effect of TZD's and has become the most frequent cause of discontinuation of therapy The incidence of TZD-mduced fluid retention ranges from 7% m monotherapy and to as high as 15% when combined with insulin (Yan, T , Soodvilai, S , PPAR Research volume 2008, article ID 943614) The mechanisms for such side-effects are not fully understood but may be related in Na and fluid re-absorption in the kidney However TZD-induced fluid retention is resistant to loop diuretics or thiazide diuretics, and combination of peroxisome prohferator- activated receptor (PPAR) alpha with PPAR gamma agonists, which were proposed to reduce such fluid overload, are associated with major adverse cardiovascular events
In view of the foregoing, it is recognized that salt and fluid accumulation contπbute to the morbidity and mortality of many diseases, including heart failure (in particular, congestive heart failure), chronic kidney disease, end-stage renal disease, liver disease and the like It is also accepted that salt and fluid accumulation are πsk factors for hypertension Accordingly, there is a clear need for a medicament that, when administered to a patient in need, would result in a reduction in sodium retention, fluid retention, or preferably both Such a medicament would more preferably also not involve or otherwise impair renal mechanisms of fluid/Na homeostasis
One option to consider for treating excessive fluid overload is to induce diarrhea Diarrhea may be triggered by several agents including, for example, laxatives such as sorbitol, polyethyleneglycol, bisacodyl and phenolphthaleme Sorbitol and polyethyleneglycol triggers osmotic diarrhea with low levels of secreted electrolytes, thus, their utility in removing sodium salt from the GI tract is limited The mechanism of action of phenolphthalem is not clearly established, but is thought to be caused by inhibition of the Na/K ATPase and the CI/HCO3 anion exchanger and stimulation of electrogenic anion secretion (see, e g , Eherer, A J , C A Santa Ana, J Porter, and J S Fordtran, 1993, Gastroenterology, v 104, no 4, p 1007-1012) However, some laxatives, such as phenolphthalem, are not viable options for the chronic treatment of fluid overload, due to the potential πsk of carcinogenicity m humans Furthermore, laxatives may not be used chronically, as they have been shown to be an irritant and cause mucosal damage Accordingly, it should also be recognized that the induction of chronic diarrhea as part of an effort to control salt and fluid overload would be an undesired treatment modality for most patients Any medicament utilizing the GI tract for this purpose would therefore need to control diarrhea in order to be of practical benefit
One approach for the treatment of mild diarrhea is the administration of a fluid-absorbmg polymer, such as the natural plant fiber psyllium Polymeric mateπals, and more specifically hydrogel polymers, may also be used for the removal of fluid from the gastrointestinal (GI) tract The use of such polymers is described m, for example, U S Pat No 4,470,975 and No 6,908,609, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes However, for such polymers to effectively remove significant quantities of fluid, they must desirably resist the static and osmotic pressure range existing in the GI tract Many mammals, including humans, make a soft feces with a water content of about 70%, and do so by transporting fluid against the high hydraulic resistance imposed by the fecal mass Several studies show that the pressure required to dehydrate feces from about 80% to about 60% is between about 500 kPa and about 1000 kPa (i e , about 5 to about 10 atm) (See, e g , McKie, A T , W Powne, and R J Naftalm, 1990, Am J Physiol, v 258, no 3 Pt 1, p G391-G394, Bleakman, D , and R J Naftalm, 1990, Am J Physiol, v 258, no 3 Pt 1, p G377-G390, Zammit, P S , M Mendizabal, and R J Naftalm, 1994, J Physiol, v 477 ( Pt 3), p 539-548 ) However, the static pressure measured intralummally is usually between about 6 kPa and about 15 kPa The rather high pressure needed to dehydrate feces is essentially due to an osmotic process and not a mechanical process produced by muscular forces The osmotic pressure aπses from the active transport of salt across the colonic mucosa that ultimately produces a hypertonic fluid absorption The osmotic gradient produced dnves fluid from the lumen to the serosal side of the mucosa Fluid-absorbing polymers, such as those described in for example U S Patent Nos 4,470,975 and 6,908,609, may not be able to sustain such pressure Such polymers may collapse in a normal colon where the salt absorption process is mtact, hence removing a modest quantity of fluid and thereby salt Synthetic polymers that bind sodium have also been descπbed For example, ion-exchange polymeric resms, such as Dowex-type cation exchange resms, have been known since about the 1950's However, with the exception of Kayexalate™
(or Kionex™), which is a polystyrene sulfonate salt approved for the treatment of hyperkalemia, cation exchange resins have very limited use as drugs, due at least in part to their limited capacity and poor cation binding selectivity Additionally, duπng the ion-exchange process, the resms may release a stochiometπc amount of exogenous cations (e g , H, K, Ca), which may in turn potentially cause acidosis (H), hyperkalemia
(K) or contπbute to vascular calcification (Ca) Such resins may also cause constipation
Gastrointestinal Tract Disorders
Constipation is characterized by infrequent and difficult passage of stool and becomes chrome when a patient suffers specified symptoms for over 12 non- consecutive weeks withm a 12-month period Chrome constipation is idiopathic if it is not caused by other diseases or by use of medications An evidence-based approach to the management of chronic constipation in North Amenca (Brandt et al , 2005, Am J Gastroenterol 100(Suρρl 1) S5-S21) revealed that prevalence is approximately 15% of the general population Constipation is reported more commonly in women, the elderly, non-whites, and individuals from lower socioeconomic groups
Irritable bowel syndrome (IBS) is a common GI disorder associated with alterations in motility, secretion and visceral sensation A range of clinical symptoms characterizes this disorder, including stool frequency and form, abdominal pam and bloating The recognition of clmical symptoms of IBS are yet to be defined, but it is now common to refer to diarrhea-predominant IBS (D-IBS) and constipation- predominant IBS (C-IBS), wherein D-IBS is defined as continuous passage of loose or watery stools and C-IBS as a group of functional disorders which present as difficult, infrequent or seemingly incomplete defecation The pathophysiology of IBS is not fully understood, and a number of mechanisms have been suggested Visceral hypersensitivity is often considered to play a major etiologic role and has been proposed to be a biological marker even useful to discriminate IBS from other causes of abdominal pam In a recent clinical study (Posserud, I et al, Gastroenterology, 2007,133 1113 1123) IBS patients were submitted to a visceral sensitivity test (Balloon distention) and compared with healthy subjects It revealed that 61% of the IBS patients had an altered visceral perception as measured by pam and discomfort threshold Other reviews have documented the role of visceral hypersensitivity in abdominal pam symptomatic of vaπous gastrointestinal tract disorders (Akbar, A, et al, Aliment Pharmaco Ther ,2009,30,423-435, Bueno et al , Neurogastroenterol Motility (2007) 19 (suppl 1), 89-119) Colonic and rectal distention have been widely used as a tool to assess visceral sensitivity m animal and human studies The type of stress used to induce visceral sensitivity vanes upon the models (see for instance Eutamen, H Neurogastroenterol Motil 2009 Aug 25 [Epub ahead of pπnt]), however stress such as Partial restraint stress (PRS) is a relatively mild, non-ulcerogenic model that is considered more representative of the IBS setting
Constipation is commonly found in the geπatnc population, particularly patients with osteoporosis who have to take calcium supplements Calcium supplements have shown to be beneficial in ostoporotic patients to restore bone density but compliance is poor because of calcium-induced constipation effects
Opioid-induced constipation (OIC) (also referred to as opioid-mduced bowel dysfunction or opioid bowel dysfuntion (OBD)) is a common adverse effect associated with opioid therapy OIC is commonly descnbed as constipation, however, it is a constellation of adverse gastrointestinal (GI) effects, which also includes abdominal cramping, bloating, and gastroesophageal reflux Patients with cancer may have disease-related constipation, which is usually worsened by opioid therapy However, OIC is not limited to cancer patients A recent survey of patients taking opioid therapy for pam of non cancer origin found that approximately 40% of patients expeπenced constipation related to opioid therapy (<3 complete bowel movements per week) compared with 7 6% in a control group Of subjects who required laxative therapy, only 46% of opioid-treated patients (control subjects, 84%) reported achieving the desired treatment results >50% of the time (Pappagallo, 2001, Am J Surg 182(5A Suppl ) 11S-18S) Some patients suffeπng from chronic idiopathic constipation can be successfully treated with lifestyle modification, dietary changes and increased fluid and fiber intake, and these treatments are generally tried first For patients who fail to respond to these approaches, physicians typically recommend laxatives, most of which are available over-the-counter Use of laxatives provided over-the-counter is judged inefficient by about half of the patients (Johanson and Kralstem, 2007, Aliment Pharmacol Ther 25(5) 599-608) Other therapeutic options currently prescπbed or in clinical development for the treatment of IBS and chronic constipation including OIC are descπbed m, for example Chang et al , 2006, Curr Teat Options Gastroenterol 9(4) 314-323, Gershon and Tack, 2007, Gastroenterology 132(1) 397-414, and, Hammerle and Surawicz, 2008, World J Gastroenterol 14(17) 2639-2649 Such treatments include but are not limited to serotonin receptor hgands, chloπde channel activators, opioid receptor antagonists, guanylate-cyclase receptor agonists and nucleotide P2Y(2) receptor agonists Many of these treatment options are inadequate, as they may be habit forming, ineffective in some patients, may cause long term adverse effects, or otherwise are less than optimal
Na+ / H+ Exchanger (NHE) Inhibitors
A major function of the GI tract is to maintain water/Na homeostasis by absorbing virtually all water and Na to which the GI tract is exposed The epithelial layer covering the apical surface of the mammalian colon is a typical electrolyte- transporting epithelium, which is able to move large quantities of salt and water in both directions across the mucosa For example, each day the GI tract processes about 9 liters of fluid and about 800 meq of Na (See, e g , Zachos et al , Molecular physiology of intestinal Na+/H+ exchange, Annu Rev Physiol , v 67, p 411-443 (2005) ) Only about 1 5 liters of this fluid and about 150 meq of this sodium originates from ingestion, rather, the majoπty of the fluid (e g , about 7 5 liters) and sodium (about 650 meq) is secreted via the GI organs as part of digestion The GI tract therefore represents a viable target for modulating systemic sodium and fluid levels Many reviews have been published on the physiology and secretory and/or absorption mechanisms of the GI tract (see, e g , Kunzelmann et al , Electrolyte transport in the mammalian colon mechanisms and implications for disease, Physiol Rev , v 82, no l, p 245-289 (2002), Geibel, J P , Secretion and absorption by colonic crypts, Annu Rev Physiol, v 67, p 471-490 (2005), Zachos et al , supra, Kiela, P R et al , Apical NA+/H+ exchangers in the mammalian gastrointestinal tract, J Physiol Pharmacol , v 57 Suppl 7, p 51-79 (2006)) The two main mechanisms of Na absorption are electroneutral and electrogemc transport Electroneutral transport is essentially due to the Na+/H+ antiport NHE (e g , NHE-3) and is responsible for the bulk of Na absorption Electrogemc transport is provided by the epithelium sodium channel ("ENaC") Electroneutral transport is located pπmaπly in the ileal segment and proximal colon and electrogemc transport is located in the distal colon
Plasma membrane NHEs contπbute to maintenance of intracellular pH and volume, transcellular absorption of NaCl and NaHCC>3, and fluid balance earned out by epithelial cells, especially in the kidney, intestine, gallbladder, and salivary glands, as well as regulation of systemic pH There exists a body of literature devoted to the role and clinical intervention on systemic NHEs to treat disorders related to ischemia and reperfusion for cardioprotection or renal protection Nine isoforms of NHEs have been identified (Kiela, P R , et al , Apical NA+/H+ exchangers in the mammalian gastrointestinal tract, J Physiol Pharmacol , v 57 Suppl 7, p 51-79 (2006)), of which NHE 2, NHE 3 and NHE 8 are expressed on the apical side of the GI tract, with NHE 3 providing a larger contribution to transport Another, yet to be identified, Cl-dependant NHE has been identified in the crypt of rat cells In addition, much research has been devoted to identifying inhibitors of NHEs The primary targets of such research have been NHE-I and NHE-3 Small molecule NHE inhibitors are, for example, descπbed in U S Patent Nos 5,866,610, 6,399,824, 6,911,453, 6,703,405, 6,005,010, 6,736,705, 6,887,870, 6,737,423, 7,326,705, 5,824,691 (WO 94/026709), 6,399,824 (WO 02/024637), U S Pat Pub Nos 2004/0039001 (WO 02/020496), 2005/0020612 (WO 03/055490), 2004/0113396 (WO 03/051866), 2005/0020612, 2005/0054705, 2008/0194621, 2007/0225323, 2004/0039001 , 2004/0224965, 2005/0113396, 2007/0135383, 2007/0135385, 2005/0244367, 2007/0270414, International Publication Nos WO 01/072742, WO 01021582 (CA2387529), WO 97/024113 (CA02241531) and European Pat No EP0744397 (CA2177007), all of which are incorporated herein by reference in their entirety for all relevant and consistent purposes However, to-date, such research has failed to develop or recognize the value or importance of NHE inhibitors that are not absorbed (i e , not systemic) and target the gastrointestinal tract Such inhibitors could be utilized in the treatment of disorders associated with fluid retention and salt overload and m the treatment of GI tract disorders, including the treatment or reduction of pain associated with a gastrointestinal tract disorder Such inhibitors would be particular advantageous because they could be delivered with reduced fear of systemic on-target or off-target effects (e g , little or no nsk of renal involvement or other systemic effects
Accordingly, while progress has been made in the foregoing fields, there remains a need in the art for novel compounds for use in the disorders associated with fluid retention and salt overload and in the treatment of gastrointestinal tract disorders, including the treatment or reduction of pam associated with a gastrointestinal tract disorder The present invention fulfills this need and provides further related advantages
BRIEF SUMMARY
In brief, the present invention is directed to compounds that are substantially active in the gastrointestinal tract to inhibit NHE-mediated antiport of sodium ions and hydrogen ions, and the use of such compounds in the treatment of disorders associated with fluid retention and salt overload and in the treatment of gastrointestinal tract disorders, including the treatment or reduction of pam associated with a gastrointestinal tract disorder In one embodiment, a compound is provided having (l) a topological
Polar Surface Area (tPSA) of at least about 200 A2 and a molecular weight of at least about 710 Daltons in the non-salt form, or (ii) a tPSA of at least about 270 A2, wherein the compound is substantially active in the gastrointestinal tract to inhibit NHE- mediated antiport of sodium ions and hydrogen ions therein upon administration to a patient in need thereof In further embodiments, the compound has a molecular weight of at least about 500 Da, at least about 1000 Da, at least about 2500 Da, or at least about about 5000 Da
In further embodiments, the compound has a tPS A of at least about 250 A2, at least about 270 A2, at least about 300 A2, at least about 350 A2, at least about 400 A2, or at least about 500 A2
In further embodiments, the compound is substantially active on the apical side of the epithelium of the gastrointestinal tract to inhibit antiport of sodium ions and hydrogen ions mediated by NHE-3, NHE-2, NHE-8, or a combination thereof In further embodiments, the compound is substantially systemically non-bioavailable and/or substantially impermeable to the epithelium of the gastrointestinal tract In farther embodiments, the compound is substantially active in the lower gastrointestinal tract In further embodiments, the compound has (i) a total number of NH and/or OH and/or other potential hydrogen bond donor moieties greater than about 5, (ii) a total number of O atoms and/or N atoms and/or other potential hydrogen bond acceptors greater than about 10, and/or (in) a Moiiguchi partition coefficient greater than about 105 or less than about 10 In further embodiments, the compound has a permeability coefficient, Papp, of less than about 100 x 106 cm/s, or less than about 10 x 106 cm/s, or less than about 1 x 106 cm/s, or less than about 0 1 x 106 cm/s In further embodiments, the compound is substantially localized in the gastrointestinal tract or lumen In further embodiments, the compound inhibits NHE irreversibly In further embodiments, the compound is capable of providing a substantially persistent inhibitory action and wherein the compound is orally administered once-a-day In further embodiments, the compound is substantially stable under physiological conditions in the gastrointestinal tract In further embodiments, the compound is inert with regard to gastrointestinal flora In further embodiments, the compound is designed to be delivered to the lower part of the gastrointestinal tract In farther embodiments, the compound is designed to be delivered to the lower part of the gastrointestinal tract past the duodenum In farther embodiments, the compound, when administered at a dose resulting in at least a 10% increase in fecal water content, has a Cmaχ that is less than the IC50 for NHE-3, less than about 1OX the IC50, or less than about IOOX the IC50 In further embodiments, upon administration of the compound to a patient m need thereof, the compound exhibits a maximum concentration detected in the serum, defined as Cmax, that is lower than the NHE inhibitory concentration IC50 of the compound In further embodiments, upon administration of the compound to a patient in need thereof, greater than about 80%, greater than about 90% or greater than about 95% of the amount of compound administered is present in the patient's feces
In further embodiments, the compound has a structure of Formula (I) or (IX)
NHE — Z (I)
NHEH — Z
J E (IX) wherem NHE is a NHE-mhibitmg small molecule that compπses (1) a hetero- atom containing moiety, and (11) a cyclic or heterocyclic scaffold or support moiety bound directly or indirectly thereto, the heteroatom-containing moiety being selected from a substituted guamdmyl moiety and a substituted heterocyclic moiety, which may optionally be fused with the scaffold or support moiety to form a fused bicyclic structure, and,
Z is a moiety having at least one site thereon for attachment to the NHE- inhibiting small molecule, the resulting NHE-Z molecule possessing overall physicochemical properties that render it substantially impermeable or substantially systemically non bioavailable, and, E is an integer having a value of 1 or more
In further embodiments, the total number of freely rotatable bonds m the
NHE-Z molecule is at least about 10 In further embodiments, the total number hydrogen bond donors in the NHE-Z molecule is at least about 5 In further embodiments, the total number of hydrogen bond acceptors in the NHE-Z molecule is at least about 10 In further embodiments, the total number of hydrogen bond donors and hydrogen bond acceptors m the NHE-Z molecule is at least about 10 In further embodiments, the Log P of the NHE-Z inhibiting compound is at least about 5 In further embodiments, the log P of the NHE-Z inhibiting compound is less than about 1, or less than about 0 In further embodiments, the scaffold is a 5-member or 6-member cyclic or heterocyclic moiety In further embodiments, the scaffold is aromatic
In further embodiments, the scaffold of the NHE-inhibitmg small molecule is bound to the moiety, Z, and the compound has the structure of Formula (II)
Substantially impermeable and/or substantially systemically non-bioavailable
NHE inhibiting compound
Figure imgf000017_0001
NHE-i nhibiting Small Molecule
(H) wherein
Z is a Core having one or more sites thereon for attachment to one or more NHE-mhibitmg small molecules, the resulting NHE-Z molecule possessing overall physicochemical properties that render it substantially impermeable or substantially systemically non-bioavailable, B is the heteroatom-containing moiety of the NHE-inhibitmg small molecule, and is selected from a substituted guanidinyl moiety and a substituted heterocyclic moiety, which may optionally be fused with the Scaffold moiety to form a fused, bicyclic structure,
Scaffold is the cyclic or heterocyclic scaffold or support moiety of the NHE-inhibitmg small molecule, which is bound directly or indirectly to heteroatom- containing moiety, B, and which is optionally substituted with one or more additionally hydrocarbyl or heterohydrocarbyl moieties,
X is a bond or a spacer moiety selected from a group consisting of substituted or unsubstituted hydrocarbyl or heterohydrocarbyl moieties, and in particular substituted or unsubstituted Ci 7 hydrocarbyl or heterohydrocarbyl, and substituted or unsubstituted, saturated or unsaturated, cyclic or heterocyclic moieties, which links B and the Scaffold, and,
D and E are integers, each independently having a value of 1 or more In further embodiments, the compound is an oligomer, dendπmer or polymer, and Z is a Core moiety having two or more sites thereon for attachment to multiple NHE-mhibitmg small molecules, either directly or indirectly through a linking moiety, L, and the compound has the structure of Formula (X)
Figure imgf000018_0001
(X)
wherein L is a bond or linker connecting the Core to the NHE-inhibitmg small molecule, and n is an integer of 2 or more, and further wherein each NHE- inhibiting small molecule may be the same or differ from the others
In further embodiments, the NHE-mhibiting small molecule has the structure of Formula (IV)
Figure imgf000018_0002
(IV) or a stereoisomer, prodrug or pharmaceutically acceptable salt thereof, wherein each Ri, R2, R3, R5 and R9 are independently selected from H, halogen, -
NR7(CO)R8, -(CO)NR7R8, -SO2-NR7R8, -NR7SO2R8, -NR7R8, -OR7, -SR7, - 0(CO)NR7R8, -NR7(CO)OR8, and -NR7SO2NR8, where R7 and R8 are independently selected from H or a bond linking the NHE-mhibitmg small molecule to L, provided at least one is a bond linking the NHE-inhibihng small molecule to L, R4 is selected from H, Ci-C7 alkyl, or a bond linking the NHE-mhibiting small molecule to L,
R.6 is absent or selected from H and Ci-C7 alkyl, and
ArI and Ar2 independently represent an aromatic πng or a heteroaromatic nng
In further embodiments, the NHE-mhibitmg small molecule has the following structure
Figure imgf000019_0001
or a stereoisomer, prodrug or pharmaceutically acceptable salt thereof, wherein each R1, R2 and R3 are independently selected from H, halogen, -
NR7(CO)R8, -(CO)NR7R8, -SO2-NR7R8, -NR7SO2R8, -NR7R8, -OR7, -SR7, -
0(CO)NR7R8, -NR7(CO)OR8, and -NR7SO2NR8, where R7 and R8 are independently selected from H or a bond linking the NHE-inhibiting small molecule to L, provided at least one is a bond linking the NHE-inhibitmg small molecule to L
In further embodiments, the NHE-inhibiting small molecule has one of the following structures
Figure imgf000019_0002
or a stereoisomer, prodrug or pharmaceutically acceptable salt thereof In further embodiments, L is a polyalkylene glycol linker In further embodiments, L is a polyethylene glycol linker In further embodiments, n is 2
In further embodiments, the Core has the following structure
!-x-γ-x-| wherein
X is selected from the group consisting of a bond, -O-, -NH-, -S-, Ci 6alkylene, -NHC(=O)-, -Q=O)NH-, -NHC(=O)NH-, -SO2NH-, and -NHSO2-, Y is selected from the group consisting of a bond, optionally substituted
Ci salkylene, optionally substituted aryl, optionally substituted heteroaryl, a polyethylene glycol linker, -(CH2)i 6θ(CH2)i 6- and -(CH2)i 6NY,(CH2)i 6-, and
Yi is selected from the group consisting of hydrogen, optionally substituted Ci salkyl, optionally substituted aryl or optionally substituted heteroaryl In further embodiments, the Core is selected from the group consisting
Figure imgf000020_0001
O OH
OH O
Figure imgf000020_0002
O OH H OH O ,
Figure imgf000021_0001
In further embodiments, the compound is an oligomer, and Z is a linking moiety, L, that links two or more NHE-mhibitmg small molecules together, when the two or more NHE-inhibiting small molecules may be the same or different, and the compound has the structure of Formula (XI)
NHE-+— L NHE-] — L
» ' m
(XI)
wherein L is a bond or linker connecting one NHE-mhibiting small molecule to another, and m is 0 or an integer of 1 or more
In further embodiments, the compound is an oligomer, dendnmer or polymer, and Z is a backbone, denoted Repeat Unit, to which is bound multiple NHE- inhibiting moieties, and the compound has the structure of Formula (XIIB)
. — [ — 1 repeat unit-) — )
\ / n (XIIB)
wherein L is a bond or a linking moiety, NHE is a NHE-inhibitmg small molecule, and n is a non-zero integer In another embodiment, a pharmaceutical composition is provided comprising a compound as set forth above, or a stereoisomer, pharmaceutically acceptable salt or prodrug thereof, and a pharmaceutically acceptable earner, diluent or excipient
In further embodiments, the composition further comprises a fluid- absorbing polymer In further embodiments, the flmd-absorbmg polymer is delivered directly to the colon In further embodiments, the fluid-absorbing polymer has a fluid absorbency of at least about 15 g of isotonic fluid per g of polymer under a static pressure of about 5 kPa In further embodiments, the fluid-absorbing polymer has a fluid absorbency of at least about 15 g of isotonic fluid per g of polymer under a static pressure of about 10 kPa In further embodiments, the fluid-absorbing polymer is characterized by a fluid absorbency of at least about 10 g/g In further embodiments, the fluid-absorbing polymer is characteπzed by a fluid absorbency of at least about 15 g/g In further embodiments, the fluid-absorbing polymer is superabsorbent In further embodiments, the fluid absorbing polymer is a crosshnked, partially neutralized polyelectrolyte hydrogel In further embodiments, the fluid-absorbing polymer is a crosshnked polyacrylate In further embodiments, the fluid-absorbing polymer is a polyelectrolyte In further embodiments, the fluid-absorbmg polymer is calcium Carbophil In further embodiments, the fluid-absorbing polymer is prepared by a high internal phase emulsion process In further embodiments, the flmd-absorbmg polymer is a foam In further embodiments, the fluid-absorbing polymer is prepared by a aqueous free radical polymerization of acrylamide or a deπvative thereof, a crosslinker and a free radical initiator redox system in water In further embodiments, the fluid- absorbing polymer is a hydrogel In further embodiments, the fluid-absorbing polymer is an N-alkyl acrylamide In further embodiments, the flmd-absorbmg polymer is a superporous gel In further embodiments, the fluid-absorbing polymer is naturally occurring In further embodiments, the fluid-absorbing polymer is selected from the group consisting of xanthan, guar, wellan, hemicelluloses, alkyl-cellulose hydro-alkyl- cellulose, carboxy-alkyl-cellulose, carrageenan, dextran, hyaluronic acid and agarose In further embodiments, the fluid-absorbing polymer is psyllium In further embodiments, the fluid-absorbmg polymer is a polysaccharide that includes xylose and arabmose In further embodiments, the fluid-absorbing polymer is a polysaccharide that includes xylose and arabmose, wherein the ratio of xylose to arabmose is at least about 3 1 , by weight
In further embodiments, the composition further comprises another pharmaceutically active agent or compound In further embodiments, the composition further compπses another pharmaceutically active agent or compound selected from the group consisting of a diuretic, cardiac glycoside, ACE inhibitor, angiotensm-2 receptor antagonist, calcium channel blocker, beta blocker, alpha blocker, central alpha agonist, vasodilator, blood thinner, anti-platelet agent, hpid-loweπng agent, and peroxisome prohferator-activated receptor (PPAR) gamma agonist agent In further embodiments, the diuretic is selected from the group consisting of a high ceiling loop diuretic, a benzothiadiazide diuretic, a potassium spaπng diuretic, and a osmotic diuretic In further embodiments, the composition further compπses another pharmaceutically active agent or compound selected from the group consisting of an analgesic peptide or agent In further embodiments, the composition further compπses another pharmaceutically active agent or compound selected from the group consisting of a laxative agent selected from a bulk-producmg agent (e g psyllium husk (Metamucil)), methylcellulose (Citrucel), polycarbophil, dietary fiber, apples, stool softeners/surfactant (e g , docusate, Colace, Diocto), a hydrating or osmotic agent (e g , dibasic sodium phosphate, magnesium citrate, magnesium hydroxide (Milk of magnesia), magnesium sulfate (which is Epsom salt), monobasic sodium phosphate, sodium biphosphate), a hyperosmotic agent (e g , glyceπn suppositoπes, sorbitol, lactulose, and polyethylene glycol (PEG))
In another embodiment, a method for inhibiting NHE-mediated antiport of sodium and hydrogen ions is provided, the method composing administering to a mammal in need thereof a pharmaceutically effective amount of a compound or pharmaceutical composition as set forth above In another embodiment, a method for treating a disorder associated with fluid retention or salt overload is provided, the method composing administering to a mammal in need thereof a pharmaceutically effective amount of a compound or pharmaceutical composition as set forth above In another embodiment, a method for treating a disorder selected from the group consisting of heart failure (such as congestive heart failure), chrome kidney disease, end-stage renal disease, liver disease, and peroxisome proliferator-activated receptor (PPAR) gamma agonist-induced fluid retention is provided, the method compπsing administering to a mammal in need thereof a pharmaceutically effective amount of a compound or pharmaceutical composition as set forth above
In another embodiment, a method for treating hypertension is provided, the method compπsing administering to a mammal in need thereof a pharmaceutically effective amount of a compound or pharmaceutical composition as set forth above
In further embodiments, the method compπses administering a pharmaceutically effective amount of the compound to the mammal in order to increase the mammal's daily fecal output of sodium and/or fluid In further embodiments, the method compπses administeπng a pharmaceutically effective amount of the compound to the mammal in order to increase the mammal's daily fecal output of sodium by at least about 30 mmol, and/or fluid by at least about 200 ml hi further embodiments, the mammal's fecal output of sodium and/or fluid is increased without introducing another type of cation m a stoichiometπc or near stoichiometπc fashion via an ion exchange process In further embodiments, the method further compπses administering to the mammal a fluid-absorbing polymer to absorb fecal fluid resulting from the use of the compound that is substantially active m the gastrointestinal tract to inhibit NHE- mediated antiport of sodium ions and hydrogen ions therein
In further embodiments, the compound or composition is administered to treat hypertension In further embodiments, the compound or composition is administered to treat hypertension associated with dietary salt intake In further embodiments, administration of the compound or composition allows the mammal to intake a more palatable diet In further embodiments, the compound or composition is administered to treat fluid overload In further embodiments, the fluid overload is associated with congestive heart failure In further embodiments, the fluid overload is associated with end stage renal disease In further embodiments, the fluid overload is associated with peroxisome prohferator-activated receptor (PPAR) gamma agonist therapy In further embodiments, the compound or composition is administered to treat sodium overload In further embodiments, the compound or composition is administered to reduce mterdialytic weight gam m ESRD patients In further embodiments, the compound or composition is administered to treat edema In further embodiments, the edema is caused by chemotherapy, pre-menstrual fluid overload or preeclampsia In further embodiments, the compound or composition is administered orally, by rectal suppository, or enema
In further embodiments, the method compπses administering a pharmaceutically effective amount of the compound or composition in combination with one or more additional pharmaceutically active compounds or agents In further embodiments, the one or more additional pharmaceutically active compounds or agents is selected from the group consisting of a diuretic, cardiac glycoside, ACE inhibitor, angiotensin-2 receptor antagonist, aldosterone antagonist, calcium channel blocker, beta blocker, alpha blocker, central alpha agonist, vasodilator, blood thinner, anti-platelet agent, hpid-loweπng agent, and peroxisome prohferator-activated receptor (PPAR) gamma agonist agent In further embodiments, the diuretic is selected from the group consisting of a high ceiling loop diuretic, a benzothiadiazide diuretic, a potassium spaπng diuretic, and a osmotic diuretic In further embodiments, the pharmaceutically effective amount of the compound or composition, and the one or more additional pharmaceutically active compounds or agents, are administered as part of a single pharmaceutical preparation In further embodiments, the pharmaceutically effective amount of the compound or composition, and the one or more additional pharmaceutically active compounds or agents, are administered as individual pharmaceutical preparations In further embodiments, the individual pharmaceutical preparation are administered sequentially In further embodiments, the individual pharmaceutical preparation are administered simultaneously In another embodiment, a method for treating a gastrointestinal tract disorder is provided, the method comprising administering to a mammal in need thereof a pharmaceutically effective amount of a compound or pharmaceutical composition as set forth above In further embodiments, the gastrointestinal tract disorder is a gastrointestinal motility disorder In further embodiments, the gastrointestinal tract disorder is irritable bowel syndrome In further embodiments, the gastrointestinal tract disorder is chronic constipation In further embodiments, the gastrointestinal tract disorder is chronic idiopathic constipation In further embodiments, the gastrointestinal tract disorder is chronic constipation occurring in cystic fibrosis patients In further embodiments, the gastrointestinal tract disorder is opioid-mduced constipation In further embodiments, the gastrointestinal tract disorder is a functional gastrointestinal tract disorder In further embodiments, the gastrointestinal tract disorder is selected from the group consisting of chronic intestinal pseudo-obstruction and colonic pseudo- obstruction In further embodiments, the gastrointestinal tract disorder is Crohn's disease In further embodiments, the gastrointestinal tract disorder is ulcerative colitis In further embodiments, the gastrointestinal tract disorder is a disease referred to as inflammatory bowel disease In further embodiments, the gastrointestinal tract disorder is associated with chronic kidney disease (stage 4 or 5) In further embodiments, the gastrointestinal tract disorder is constipation induced by calcium supplement In further embodiments, the gastrointestinal tract disorder is constipation, and the constipation to be treated is associated with the use of a therapeutic agent In further embodiments, the gastrointestinal tract disorder is constipation, and the constipation to be treated is associated with a neuropathic disorder In further embodiments, the gastrointestinal tract disorder is constipation, and the constipation to be treated is post-surgical constipation (postoperative ileus) In further embodiments, the gastrointestinal tract disorder is constipation, and the constipation to be treated is idiopathic (functional constipation or slow transit constipation) In further embodiments, the gastrointestinal tract disorder is constipation, and the constipation to be treated is associated with neuropathic, metabolic or an endocrine disorder (e g , diabetes melhtus, renal failure, hypothyroidism, hyperthyroidism, hypocalcaemia, Multiple Sclerosis, Parkinson's disease, spinal cord lesions, neurofibromatosis, autonomic neuropathy, Chagas disease, Hirschsprung's disease or cystic fibrosis, and the like) In further embodiments, the gastrointestinal tract disorder is constipation, and the constipation to be treated is due the use of drugs selected from analgesics (e g , opioids), antihypertensives, 5 anticonvulsants, antidepressants, antispasmodics and antipsychotics
In another embodiment, a method for treating irritable bowel syndrome is provided, the method composing administering to a mammal in need thereof a pharmaceutically effective amount of an NHE-3 inhibitor compound or a pharmaceutical composition composing an NHE-3 inhibitor compound In further0 embodiments, the NHE-3 inhibitor compound or the pharmaceutical composition composing an NHE-3 inhibitor compound is a compound or pharmaceutical composition as set forth above
In further embodiments of the above embodiments, the compound or composition is administered to treat or reduce pain associated with a gastrointestinal5 tract disorder In further embodiments, the compound or composition is administered to treat or reduce visceral hypersensitivity associated with a gastrointestinal tract disorder In further embodiments, the compound or composition is administered to treat or reduce inflammation of the gastrointestinal tract In further embodiments, the compound or composition is administered to reduce gastrointestinal transit time 0 In further embodiments, the compound or composition is administered either orally or by rectal suppository
In further embodiments, the method composes admmisteong a pharmaceutically effective amount of the compound or composition, in combination with one or more additional pharmaceutically active compounds or agents In furtherS embodiments, the one or more additional pharmaceutically active agents or compounds are an analgesic peptide or agent In further embodiments, the one or more additional pharmaceutically active agents or compounds are selected from the group consisting of a laxative agent selected from a bulk-producing agent (e g psyllium husk (Metamucil)), methylcellulose (Citrucel), polycarbophil, dietary fiber, apples, stool0 softeners/surfactant (e g , docusate, Colace, Diocto), a hydrating or osmotic agent (e g , dibasic sodium phosphate, magnesium citrate, magnesium hydroxide (Milk of magnesia), magnesium sulfate (which is Epsom salt), monobasic sodium phosphate, sodium biphosphate), and a hyperosmotic agent (e g , glyceπn suppositoπes, sorbitol, lactulose, and polyethylene glycol (PEG)) In further embodiments, the pharmaceutically effective amount of the compound or composition, and the one or more additional pharmaceutically active compounds or agents, are administered as part of a single pharmaceutical preparation In further embodiments, the pharmaceutically effective amount of the compound or composition, and the one or more additional pharmaceutically active compounds or agents, are administered as individual pharmaceutical preparations In further embodiments, the individual pharmaceutical preparation are administered sequentially In further embodiments, the individual pharmaceutical preparation are administered simultaneously
These and other aspects of the invention will be apparent upon reference to the following detailed descπption
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 is a graph that illustrates the relationship between tPSA and
Permeability (Papp, as measured m the PAMPA assay) of certain example compounds, as further discussed in the Examples (under the subheading "2 Pharmacological Test
Example 2") Figures 2A and 2B are graphs that illustrate the cecum and colon water content after oral administration of certain example compounds, as further discussed in the Examples (under the subheading "3 Pharmacological Test Example 3")
Figures 3A and 3B are graphs that illustrate the dose dependent decrease of uπnary salt levels after administration of certain example compounds, as further discussed in the Examples (under the subheading "14 Pharmacological Test Example
14")
Figure 4 is a graph that illustrates a dose dependent increase in fecal water content after administration of a certain example compound, as further discussed in the Examples (under the subheading "15 Pharmacological Test Example 15") Figures 5A, 5B and 5C are graphs that illustrate that supplementing the diet with Psyllium results in a slight reduction of fecal stool form, but without impacting the ability of a certain example compound to increase fecal water content or decrease uπnary sodium, as further discussed in the Examples (under the subheading "16 Pharmacological Test Example 16")
Figure 6 is a graph that illustrates that inhibition of NHE-3 reduces hypersensitivity to distention, as further discussed in the Examples (under the subheading "17 Pharmacological Test Example 17")
Figures 7A and 7B are graphs that illustrate that inhibition of NHE-3 increases the amount of sodium excreted in feces, as further discussed in the Examples (under subheading "18 Pharmacological Test Example 18")
DETAILED DESCRIPTION
In accordance with the present disclosure, and as further detailed herein below, it has been found that the inhibition of NHE-mediated antiport of sodium ions (Na+) and hydrogen ions (H+) in the gastrointestinal tract, and more particularly the gastrointestinal epitheha, is a powerful approach to the treatment of vaπous disorders that may be associated with or caused by fluid retention and/or salt overload, and/or disorders such as heart failure (in particular, congestive heart failure), chrome kidney disease, end-stage renal disease, liver disease, and/or peroxisome prohferator-activated receptor (PPAR) gamma agomst-mduced fluid retention More specifically, it has been found that the inhibition of the NHE-mediated antiport of sodium ions and hydrogen ions in the GI tract increases the fecal excretion of sodium, effectively reducing systemic levels of sodium and fluid This, in turn, improves the clinical status of a patient suffeπng from, for example, CHF, ESRD/CKD and/or liver disease It has further been found that such a treatment may optionally be enhanced by the co administration of other beneficial compounds or compositions, such as for example a fluid-absorbing polymer The fluid-absorbing polymer may optimally be chosen so that it does not block or otherwise negatively interfere with the mechanism of action of the co-dosed NHE inhibitor
Additionally, and also as further detailed herein below, it has further been found that the inhibition of NHE-mediated antiport of sodium ions (Na+) and hydrogen ions (H+) in the gastrointestinal tract, and more particularly the gastrointestinal epithelia, is a powerful approach to the treatment of hypertension, that may be associated with or caused by fluid retention and/or salt overload More specifically, it has been found that the inhibition of the NHE-mediated antiport of sodium ions and hydrogen ions in the GI tract increases the fecal excretion of sodium, effectively reducing systemic levels of sodium and fluid This, in turn, improves the clinical status of a patient suffeπng from hypertension Such a treatment may optionally be enhanced by the co-admimstration of other beneficial compounds or compositions, such as for example a fluid-absorbing polymer The fluid-absorbmg polymer may optimally be chosen so that it does not block or otherwise negatively interfere with the mechanism of action of the co-dosed NHE inhibitor and/or hypertension
Additionally, and also as further detailed herein below, it has further been found that the inhibition of NHE-mediated antiport of sodium ions (Na+) and hydrogen ions (H+) in the gastrointestinal tract, and more particularly the gastrointestinal epithelia, is a powerful approach to the treatment of vaπous gastrointestinal tract disorders, including the treatment or reduction of pam associated with gastrointestinal tract disorders, and more particularly to the restoration of appropnate fluid secretion in the gut and the improvement of pathological conditions encountered m constipation states Applicants have further recognized that by blocking sodium ion re-absorption, the compound of the invention restore fluid homeostasis in the GI tract, particularly in situations wherein fluid secretion/absorption is altered in such a way that it results in a high degree of feces dehydration, low gut motility, and/or a slow transit-time producing constipation states and GI discomfort generally It has further been found that such a treatment may optionally be enhanced by the co- administration of other beneficial compounds or compositions, such as for example a fluid-absorbmg polymer The fluid-absorbing polymer may optimally be chosen so that it does not block or otherwise negatively interfere with the mechanism of action of the co-dosed NHE inhibitor
Due to the presence of NHEs in other organs or tissues in the body, the method of the present disclosure employs the use of compounds and compositions that are desirably highly selective or localized, thus acting substantially in the gastrointestinal tract without exposure to other tissues or organs In this way, any systemic effects can be minimized (whether they are on-target or off-target) Accordingly, it is to be noted that, as used herein, and as further detailed elsewhere herein, "substantially active in the gastrointestinal tract" generally refers to compounds 5 that are substantially systemically non-bioavailable and/or substantially impermeable to the layer of epithelial cells, and more specifically epithelium of the GI tract It is to be further noted that, as used herein, and as further detailed elsewhere herein, "substantially impermeable" more particularly encompasses compounds that are impermeable to the layer of epithelial cells, and more specifically the gastrointestinal0 epithelium (or epithelial layer) "Gastrointestinal epithelium" refers to the membranous tissue coveπng the internal surface of the gastrointestinal tract Accordingly, by being substantially impermeable, a compound has very limited ability to be transferred across the gastrointestinal epithelium, and thus contact other internal organs (e g , the brain, heart, liver, etc ) The typical mechanism by which a compound can be transferred5 across the gastrointestinal epithelium is by either transcellular transit (a substance travels through the cell, mediated by either passive or active transport passing through both the apical and basolateral membranes) and/or by paracellular transit, where a substance travels between cells of an epithelium, usually through highly restπctive structures known as "tight junctions" 0 The compounds of the present disclosure may therefore not be absorbed, and are thus essentially not systemically bioavailable at all (e g , impermeable to the gastrointestinal epithelium at all), or they show no detectable concentration of the compound m serum Alternatively, the compounds may (i) exhibit some detectable permeability to the layer of epithelial cells, and more particularly the epithelium of theS GI tract, of less than about 20% of the administered compound (e g , less than about 15%, about 10%, or even about 5%, and for example greater than about 0 5%, or 1%), but then are rapidly cleared in the liver (i e , hepatic extraction) via first-pass metabolism, and/or (ii) exhibit some detectable permeability to the layer of epithelial cells, and more particularly the epithelium of the GI tract, of less than about 20% of the0 administered compound (e g , less than about 15%, about 10%, or even about 5%, and for example greater than about 0 5%, or 1%), but then are rapidly cleared in the kidney (i e , renal excretion)
In this regard it is to be still further noted that, as used herein, "substantially systemically non-bioavailable" generally refers to the inability to detect a compound in the systemic circulation of an animal or human following an oral dose of the compound For a compound to be bioavailable, it must be transferred across the gastrointestinal epithelium (that is, substantially permeable as defined above), be transported via the portal circulation to the liver, avoid substantial metabolism in the liver, and then be transferred into systemic circulation As further detailed elsewhere herein, small molecules exhibiting an inhibitory effect on NHE-mediated antiport of sodium and hydrogen ions descπbed herein may be modified or functionahzed to render them "substantially active" in the GI tract (or "substantially impermeable" to the GI tract and/or "substantially systemically non-bioavailable" from the GI tract) by, for example, ensunng that the final compound has (i) a molecular weight of greater than about 500 Daltons (Da) (e g , greater than about 1000 Da, about 2500 Da, about 5000 Da, or even about 10000 Da) in its non-salt form, and/or (ii) at least about 10 freely rotatable bonds therein (e g , about 10, about 15 or even about 20), and/or (in) a Moπguchi Partition Coefficient of at least about 105 (or log P of at least about 5), by for example increasing the hydrophobicity of the compound (e g , inserting or installing a hydrocarbon chain of a sufficient or suitable length therein), or alternatively a Moπguchi Partition Coefficient of less than 10 (or alternatively a log P of less than about 1, or less than about 0), and/or (iv) a number of hydrogen-bond donors therein greater than about 5, about 10, or about 15, and/or (v) a number of hydrogen-bond acceptors therein greater than about 5, about 10, or about 15, and/or (vi) a total number of hydrogen-bond donors and acceptors therein of greater than about 5, about 10, or about 15, and/or, (vii) a topological polar surface area (tPSA) therein of greater than about 100 A2, about 120 A2, about 130 A2, or about 140 A2, and in some instances about 150 A2, about 200 A2, about 250 A2, about 270 A2, about 300 A2, about 400 A2, or even about 500 A2, by for example inserting or installing a sufficiently hydrophilic functional group therein (e g , a polyalkylene ether or a polyol or an lomzable group, such as a phosphonate, sulfonate, carboxylate, amine, quaternary amine, etc ), the hydrogen-bond donors/acceptor groups also contributing to compound tPSA
One or more of the above-noted methods for structurally modifying or functionahzing the NHE-mhibiting small molecule may be utilized in order to prepare a compound suitable for use in the methods of the present disclosure, so as to render the compound substantially impermeable or substantially systemically non-bioavailable, that is, one or more of the noted exemplary physical properties may be "engineered" into the NHE-mhibiting small molecule to render the resulting compound substantially impermeable or substantially systemically non-bioavailable, or more generally substantially active, in the GI tract, while still possessing a region or moiety therein that is active to inhibit NHE-mediated antiport of sodium ions and hydrogen ions
Without being being held to any particular theory, the NHE-inhibitors (e g , NHE-3, -2 and/or -8) of the instant disclosure are believed to act via a distinct and unique mechanism, causing the retention of fluid and ions in the GI tract (and stimulating fecal excretion) rather than stimulating increased secretion of said fluid and ions For example, lubiprostone (Amitiza® Sucampo/Takeda) is a bicyclic fatty acid prostaglandin El analog that activates the Type 2 Chloπde Channel (ClC-2) and increases chlonde-nch fluid secretion from the serosal to the mucosal side of the GI tract (see, e g , Pharmacological Reviews for Amitiza®, NDA package) Lmaclotide (MD-1100 acetate, Microbia/Forest Labs) is a 14 amino acid peptide analogue of an endogenous hormone, guanyhn, and indirectly activates the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) thereby inducing fluid and electrolyte secretion into the GI (see, e g , Li et al , J Exp Med , vol 202 (2005), pp 975-986) The substantially impermeable NHE inhibitors descπbed in the instant disclosure act to inhibit the reuptake of salt and fluid rather than promote secretion Since the GI tract processes about 9 liters of fluid and about 800 meq of Na each day, it is anticipated that NHE inhibition could permit the removal of substantial quantities of systemic fluid and sodium to resorb edema and resolve CHF symptoms
I. Substantially Impermeable or Substantially Systemically Non-Bioavailable NHE-Inhibiting Compounds A. General Structure
Generally speaking, the present disclosure encompasses essentially any small molecule, which may be monovalent or polyvalent, that is effective or active as a
NHE inhibitor and that is substantially active in the GI tract, and more particularly substantially impermeable or substantially systemically non-bioavalable therein, including known NHE inhibitors that may be modified or functionalized in accordance with the present disclosure to alter the physicochemical properties thereof so as to render the overall compound substantially active in the GI tract In particular, however, the present disclosure encompasses monovalent or polyvalent compounds that are effective or active as NHE-3 , NHE-2 and/or NHE-8 inhibitors
Accordingly, the compounds of the present disclosure may be generally represented by Formula (I)
NHE- Z
(I) wherein (i) NHE represents a NHE-inhibitmg small molecule, and (ii) Z represents a moiety having at least one site thereon for attachment to an NHE-mhibitmg small molecule, the resulting NHE-Z molecule possessing overall physicochemical properties that render it substantially impermeable or substantially systemically non-bioavailable The NHE-inhibitmg small molecule generally compπses a heteroatom-contaimng moiety and a cyclic or heterocyclic scaffold or support moiety bound directly or indirectly thereto In particular, examination of the structures of small molecules reported to-date to be NHE inhibitors suggest, as further illustrated herein below, that most compπse a cyclic or heterocyclic support or scaffold bound directly or indirectly (by, for example, an acyl moiety or a hydrocabyl or heterohydrocarbyl moiety, such as an alkyl, an alkenyl, a heteroalkyl or a heteroalkenyl moiety) to a heteroatom- contaimng moiety that is capable of acting as a sodium atom or sodium ion mimic, which is typically selected from a substituted guamdinyl moiety and a substituted heterocyclic moiety (e g , a mtrogen-contammg herocyclic moiety) Optionally, the heteroatom-contaimng moiety may be fused with the scaffold or support moiety to form a fused, bicyclic structure, and/or it may be capable of forming a positive charge at a physiological pH In this regard it is to be noted that, while the heteroatom-contaimng moiety that is capable of acting as a sodium atom or ion mimic may optionally form a positive charge, this should not be understood or interpreted to require that the overall compound have a net positive charge, or only a single positively charged moiety 5 therein Rather, m various embodiments, the compound may have no charged moieties, or it may have multiple charged moieties therein (which may have positive charges, negative charges, or a combination thereof, the compound for example being a zwittenon) Additionally, it is to be understood that the overall compound may have a net neutral charge, a net positive charge (e g , +1, +2, +3, etc ), or a net negative charge0 (e g , -1, -2, -3, etc )
The Z moiety may be bound to essentially any position on, or within, the NHE small molecule, and in particular may be (i) bound to the scaffold or support moiety, (ii) bound to a position on, or withm, the heteroatom-contaimng moiety, and/or (m) bound to a position on, or within, a spacer moiety that links the scaffold to the5 heteroatom-contaimng moiety, provided that the installation of the Z moiety does not significantly adversely impact NHE-inhibitmg activity In one particular embodiment, Z may be in the form of an oligomer, dendπmer or polymer bound to the NHE small molecule (e g , bound for example to the scaffold or the spacer moiety), or alternatively Z may be in the form of a linker that links multiple NHE small molecules together, and0 therefore that acts to increase (i) the overall molecular weight and/or polar surface area of the NHE-Z molecule, and/or, (ii) the number of freely rotatable bonds m the NHE-Z molecule, and/or, (in) the number of hydrogen-bond donors and/or acceptors in the NHE-Z molecule, and/or, (iv) the Log P value of the NHE-Z molecule to a value of at least about 5 (or alternatively less than 1, or even about 0), all as set forth herein, suchS that the overall NHE-mhibiting compound (i e , the NHE-Z compound) is substantially impermeable or substantially systemically non-bioavailable
The present disclosure is more particularly directed to such a substantially impermeable or substantially systemically non-bioavailable, NHE- mhibiting compound, or a pharmaceutical salt thereof, wherein the compound has the0 structure of Formula (II) Substantially Impermeable and/or substantially systemically non-bioavailable
NHE inhibiting compound
Figure imgf000036_0001
NHE-inhibiting Small Molecule
(II)
wherein (i) Z, as previously defined above, is a moiety bound to or incorporated in the NHE-inhibiting small molecule, such that the resulting NHE-Z molecule possesses overall physicochemical properties that render it substantially impermeable or substantially systemically non-bioavailable , (11) B is the heteroatom-contaimng moiety of the NHE-inhibiting small molecule, and m one particular embodiment is selected from a substituted guamdinyl moiety and a substituted heterocyclic moiety, which may optionally be fused with the Scaffold moiety to form a fused, bicychc structure, (m) Scaffold is the cyclic or heterocyclic moiety to which is bound directly or indirectly the hetero-atom containing moiety (e g , the substituted guamdinyl moiety or a substituted heterocyclic moiety), B, and which is optionally substituted with one or more additionally hydrocarbyl or heterohydrocarbyl moieties, (iv) X is a bond or a spacer moiety selected from a group consisting of substituted or unsubstituted hydrocarbyl or heterohydrocarbyl moieties, and in particular substituted or unsubstituted Cj-C7 hydrocarbyl or heterohydrocarbyl (e g , Cj-C7 alkyl, alkenyl, heteroalkyl or heteroalkenyl), and substituted or unsubstituted, saturated or unsaturated, cyclic or heterocyclic moieties (e g , G)-C7 cyclic or heterocyclic moieties), which links B and the Scaffold, and, (v) D and E are integers, each independently having a value of 1, 2 or more
In one or more particular embodiments, as further illustrated herein below, B may be selected from a guamdinyl moiety or a moiety that is a guamdinyl bioisostere selected from the group consisting of substituted cyclobutenedione, substituted imidazole, substituted thiazole, substituted oxadiazole, substituted pyrazole, or a substituted amine More particularly, B may be selected from guamdinyl, acylguanidinyl, sulfonylguamdinyl, or a guamdine bioisostere such as a cyclobutenedione, a substituted or unsubstituted 5- or 6-member heterocycle such as substituted or unsubstituted imidazole, aminoimidazole, alkyhmidizole, thiazole, oxadiazole, pyrazole, alkylthioimidazole, or other functionality that may optionally become positively charged or function as a sodium mimetic, including amines (e g , tertiary amines), alkylamines, and the like, at a physiological pH In one particularly preferred embodiment, B is a substituted guamdinyl moiety or a substituted heterocyclic moiety that may optionally become positively charged at a physiological pH to function as a sodium mimetic In one exemplary embodiment, the compound of the present disclosure (or more particularly the pharmaceutically acceptable HCl salt thereof, as illustrated) may have the structure of Formula (III)
Scaffold
Figure imgf000037_0001
NHE Inhibiting Small Molecule
Figure imgf000037_0002
Substantially Impermeable and/or substantially systemically non-bioavailable NHE Inhibiting Compound
(III) wherein Z may be optionally attached to any one of a number of sites on the NHE- inhibiting small molecule, and further wherein the Ri, R2 and R3 substituents on the aromatic πngs are as detailed elsewhere herein, and/or in U S Pat No 6,399,824, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes In this regard it is to be noted, however, that the substantially impermeable or substantially systemically non-bioavailable NHE-mhibiting compounds of the present disclosure may have a structure other than illustrated above, without departing from the scope of the present disclosure For example, m various alternative embodiments, one or both of the terminal nitrogen atoms in the guamdme moiety may be substituted with one or more substituents, and/or the modifying or functionalizing moiety Z may be attached to the NHE-mhibitmg compound by means of (i) the Scaffold, (11) the spacer X, or (in) the heteroatom-contaimng moiety, B, as further illustrated generally in the structures provided below
HCI
Figure imgf000038_0001
Scaffold HCI
Figure imgf000038_0002
In this regard it is to be further noted that, as used herein, "bioisostere" generally refers to a moiety with similar physical and chemical properties to a guamdme moiety, which in turn imparts biological properties to that given moiety similar to, again, a guamdme moiety, in this instance (See, for example, Ahmad, S et al , Ammoimidazoles as Bioisosteres of Acylguanidines Novel, Potent, Selective and Orally Bioavailable Inhibitors of the Sodium Hydrogen Exchanger Isoform-1, Boorganic & Med Chem Lett , pp 177-180 (2004), the entire contents of which is incorporated herein by reference for all relevant and consistent purposes )
As further detailed below, known NHE-mhibiting small molecules or chemotypes that may serve as suitable starting mateπals (for modification or functionahzation, in order to render the small molecules substantially impermeable or substantially systemically non-bioavailable, and/or used in pharmaceutical preparations in combination with, for example, a fluid-absorbing polymer) may generally be organized into a number of subsets, such as for example
κ Λ N^NH2
Figure imgf000039_0001
Beπzoylguanidines Heteroaroylguaπidines 'Spacer-Stretched Non Acyl Guanidines
Aroylguamdines
Figure imgf000039_0002
wherein the terminal πng (or, m the case of the non-acyl guamdmes, "R"), represent the scaffold or support moiety, the guamdme moiety (or the substituted heterocycle, and more specifically the pipeπdine πng, in the case of the non-guamdine inhibitors) represents B, and, X is the acyl moiety, or the -A-B-acyl- moiety (or a bond in the case of the non-acyl guanidmes and the non-guamdine inhibitors) (See, e g , Lang, H J , "Chemistry of NHE Inhibitors ' in The Sodium-Hydrogen Exchanger, Harmazyn, M , Avkiran, M and Fliegel, L , Eds , Kluwer Academic Publishers 2003 See also B Masereel et al , An Overview of Inhibitors of Na+ / H+ Exchanger, European J of Med Chem , 38, pp 547-554 (2003), the entire contents of which is incorporated by reference here for all relevant and consistent purposes) Without being held to any particular theory, it has been proposed that a guamdine group, or an acylguanidme group, or a charged guamdme or acylguanidme group (or, in the case of non-guamdine inhibitors, a heterocycle or other functional group that can replicate the molecular interactions of a guanidinyl functionality including, but not limited to, a protonated nitrogen atom in a pipendine πng) at physiological pH may mimic a sodium ion at the binding site of the exchanger or antiporter (See, e g , Vigne, P , Frelm, C , Lazdunski, M J Biol Chem 1982, 257, 9394)
Although the heteroatom-containing moiety may be capable of forming a positive charge, this should not be understood or interpreted to require that the overall compound have a net positive charge, or only a single positively charged moiety therein, or even that the heteroatom-contaimng moiety therein be capable of forming a positive charge in all instances Rather, in various alternative embodiments, the compound may have no charged moieties therein, or it may have multiple charged 5 moieties therein (which may have positive charges, negative charges, or a combination thereof) Additionally, it is to be understood that the overall compound may have a net neutral charge, a net positive charge, or a net negative charge
In this regard it is to be noted that the U S Patents and U S Published Applications cited above, or elsewhere herein, are incorporated herein by reference in
10 their entirety, for all relevant and consistent purposes
In addition to the structures illustrated above, and elsewhere herein, it is to be noted that bioisostenc replacements for guamdine or acylguanidine may also be used Potentially viable bioisostenc "guamdine replacements" identified to-date have a five- or six-membered heterocyclic πng with donor/acceptor and pKa patterns similar to
I5 that of guamdine or acylguanidine (see for example Ahmad, S et al , Ammoimidazoles as Bioisosteres of Acylguanidines Novel, Potent, Selective and Orally Bioavailable Inhibitors of the Sodium Hydrogen Exchanger Isoform-1, Boorganic & Med Chem Lett , pp 177-180 (2004), the entire contents of which is incorporated herein by reference for all relevant and consistent purposes), and include those illustrated below 20
-,.-"-/ R2 Examples of acyl i X κ m 9uanidine ιsosteres
) NH2
Λylguanidine or sodium bioisostere
Figure imgf000040_0001
The above bioisostenc embodiments (i e , the group of structures above) correspond to "B" in the structure of Formula (II), the broken bond therein being attached to "X" (e g , the acyl moiety, or alternatively a bond linking the bioisostere to the scaffold), with bonds to Z m Formula (III) not shown here It is to be noted that, m the many structures illustrated herein, all of the various linkages or bonds will not be shown m every instance For example, in one or more of the structures illustrated above, a bond or connection between the NHE- mhibiting small molecule and the modifying or functionahzmg moiety Z is not always shown However, this should not be viewed in a limiting sense Rather, it is to be understood that the NHE-mhibiting small molecule is bound or connected in some way (e g , by a bond or linker of some kind) to Z, such that the resulting NHE-Z molecule is suitable for use (i e , substantially impermeable or substantially systemically non- bioavailable in the GI tract) Alternatively, Z may be incorporated into the NHE- inhibitmg small molecule, such as for example by positioning it between the guamdme moiety and scaffold
It is to be further noted that a number of structures are provided herein for substantially impermeable or substantially systemically non-bioavailable NHE- mhibitmg compounds, and/or for NHE-mhibitmg small molecules suitable for modification or functionalization in accordance with the present disclosure so as to render them substantially impermeable or substantially systemically non-bioavailable Due to the large number of structures, various identifiers (e g , atom identifiers in a chain or nng, identifiers for substituents on a πng or chain, etc ) may be used more than once An identifier in one structure should therefore not be assumed to have the same meaning in a different structure, unless specifically stated (e g , "Ri" in one structure may or may not be the same as "Ri" in another structure) Additionally, it is to be noted that, in one or more of the structures further illustrated herein below, specific details of the structures, including one or more of the identifiers therein, may be provided m a cited reference, the contents of which are specifically incorporated herein by reference for all relevant and consistent purposes
B. Illustrative Small Molecule Embodiments The substantially impermeable or substantially systemically non- bioavailable NHE-inhibiting compounds of the present disclosure may in general be deπved or prepared from essentially any small molecule possessing the ability to inhibit NHE activity, including small molecules that have already been reported or identified as inhibiting NHE activity but lack impermeability (i e , are not substantially impermeable) In one particularly preferred embodiment, the compounds utilized in the various methods of the present disclosure are deπved or prepared from small molecules that inhibit the NHE-3, -2, and/or -8 isoforms To-date, a considerable amount of work has been devoted to the study of small molecules exhibiting NHE-I inhibition, while less has been devoted for example to the study of small molecules exhibiting NHE-3 inhibition Although the present disclosure is directed generally to substantially impermeable or substantially systemically non-bioavailable NHE-inhibiting compounds, the substantially impermeable or substantially systemically non- bioavailable compounds exhibiting NHE-3, -2, and/or -8 inhibition are of particular interest However, while it is envisioned that appropπate starting points may be the modification of known NHE-3, -2, and/or -8 inhibiting small molecules, small molecules identified for the inhibition of other NHE subtypes, including NHE-I, may also be of interest, and may be optimized for selectivity and potency for the NHE-3, -2, and/or -8 subtype antiporter Small molecules suitable for use (i e , suitable for modification or functionahzation in accordance with the present disclosure) to prepare the substantially impermeable or substantially systemically non-bioavailable NHE-inhibiting compounds of the present disclosure include those illustrated below In this regard it is to be noted a bond or link to Z (i e , the modification or functionahzation that renders the small molecules substantially impermeable or substantially systemically non-bioavailable) is not specifically shown As previously noted, the Z moiety may be attached to, or included within, the small molecule at essentially any site or position that does not interfere (e g , stencly interfere) with the ability of the resulting compound to effectively inhibit the NHE antiport of interest More particularly, Z may be attached to essentially any site on the NHE-mhibitmg small molecule, Z for example displacing all or a portion of a substituent initially or originally present thereon and as illustrated below, provided that the site of installation of the Z moiety does not have a substantially adversely impact on the NHE-inhibitmg activity thereof In one particular embodiment, however, a bond or link extends from Z to a site on the small molecule that effectively positions the point of attachment as far away (based, for example, on the number of intervening atoms or bonds) from the atom or atoms present in the resulting compound that effectively act as the sodium ion mimic (for example, the atom or atoms capable of forming a positive ion under physiological pH conditions) In a preferred embodiment, the bond or link will extend from Z to a site in a πng, and more preferably an aromatic πng, withm the small molecule, which serves as the scaffold In view of the foregoing, m one particular embodiment, the following small molecule, disclosed in U S Patent Application No 2005/0054705, the entire content of which (and in particular the text of pages 1-2 therein) is incorporated herein by reference for all relevant and consistent purposes, may be suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
Figure imgf000043_0001
The variables in the structure are defined in the cited patent application, the details of which are incorporated herein by reference In one particularly preferred embodiment, R.6 and R7 are a halogen (e g , Cl), Rs is lower alkyl (e g , CH3), and R1-R4 are H, the compound having for example the structure
Figure imgf000043_0002
In yet another particular embodiment, the following small molecule, disclosed in Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and in particular pages 1-2 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification m accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
Figure imgf000044_0001
The vaπables in the structure are defined in the cited patent application, the details of which are incorporated herein by reference
In yet another particular embodiment, the following small molecule, disclosed in Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and in particular page 49 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification m accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non bioavailable)
Figure imgf000044_0002
The vanables m the structure are defined in the cited patent application, the details of which are incorporated herein by reference In yet another particular embodiment, the following small molecule, disclosed in Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and in particular pages 118-120 and 175-177 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
R>SV R4 Y O NΥ NHN 2 H2
The variables m the structure are defined in the cited patent application, the details of which are incorporated herein by reference In yet another particular embodiment, the following small molecule, disclosed m Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and in particular pages 129-131 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification m accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
χANJ-γN γ N Hj
O NH2
The variables in the structure are defined in the cited patent application, the details of which are incorporated herein by reference (In this regard it is to be noted that the substituent Z within the structure illustrated above is not to be confused with the moiety Z that, in accordance with the present disclosure, is attached to the NHE-mhibiting small molecule m order effective render the resulting "NHE-Z" molecule substantially impermeable )
In yet another particular embodiment, the following small molecule, disclosed m Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and m particular pages 127-129 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
Figure imgf000046_0001
The variables m the structure are defined in the cited patent application, the details of which are incorporated herein by reference (In this regard it is to be noted that Z withm the nng of the structure illustrated above is not to be confused with the moiety Z that, in accordance with the present disclosure, is attached to the NHE-mhibiting small molecule m order effective render the resulting "NHE-Z" molecule substantially impermeable )
In yet another particular embodiment, the following small molecule, disclosed in Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and in particular pages 134-137 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification m accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
Figure imgf000046_0002
The variables in the structure are defined in the cited patent application, the details of which are incorporated herein by reference In yet another particular embodiment, the following small molecule, disclosed in Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/241 13), the entire content of which (and m particular pages 31- 32 and 137-139 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
Figure imgf000047_0001
The vanables in the structure are defined in the cited patent application, the details of which are incorporated herein by reference
In yet another particular embodiment, the following small molecule, disclosed in Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and in particular pages 37- 45 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
Figure imgf000047_0002
The vanables m the structure are defined in the cited patent application, the details of which are incorporated herein by reference (In this regard it is to be noted that Z within the πng structure illustrated above is not to be confused with the moiety Z that, in accordance with the present disclosure, is attached to the NHE inhibiting small molecule in order effective render the resulting "NHE-Z" molecule substantially impermeable ) In yet another particular embodiment, the following small molecule, disclosed in Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and in particular pages 100-102 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification m accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
Figure imgf000048_0001
The variables in the structure are defined in the cited patent application, the details of which are incorporated herein by reference (wherein, in particular, the wavy bonds indicate vanable length, or a vanable number of atoms, therein)
In yet another particular embodiment, the following small molecule, disclosed in Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and in particular pages 90- 91 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
Figure imgf000048_0002
The variables in the structure are defined in the cited patent application, the details of which are incorporated herein by reference
In yet another particular embodiment, the following small molecule, disclosed in U S Patent No 5,900,436 (or EP 0822182 Bl), the entire contents of which (and in particular column 1, lines 10-55 therein) are incorporated herein by reference for all relevant and consistent purposes, may be suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or 5 substantially systemically non-bioavailable)
Figure imgf000049_0001
The vaπables in the structures are defined in the cited patents, the details of which are C incorporated herein by reference
In yet another particular embodiment, the following small molecule, disclosed in Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and in particular pages 35- 47 therein) is incorporated herein for all relevant and consistent purposes, may be5 suitable for use or modification m accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
Figure imgf000049_0002
0
The vaπables in the structure are defined in the cited patent application, the details of which are incorporated herein by reference
In yet another particular embodiment, the following small molecule, disclosed in Canadian Patent Application No 2,241,531 (or International Patent5 Publication No WO 97/24113), the entire content of which (and in particular pages
154-155 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification m accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
Figure imgf000050_0001
The variables m the structure are defined in the cited patent application, the details of which are incorporated herein by reference
In yet another particular embodiment, the following small molecule, disclosed in Canadian Patent Application No 2,241,531 (or International Patent 10 Publication No WO 97/24113), the entire content of which (and in particular pages
132-133 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable) I5
Figure imgf000050_0002
The vaπables in the structure are defined m the cited patent application, the details of which are incorporated herein by reference
20 In yet another particular embodiment, the following small molecule, disclosed in Canadian Patent Application No 2,241,531 (or International Patent Publication No WO 97/24113), the entire content of which (and in particular pages 58- 65 AND 141-148 therein) is incorporated herein for all relevant and consistent purposes, may be suitable for use or modification m accordance with the present
25 disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
Figure imgf000051_0001
The variables in the structure are defined in the cited patent application, the details of which are incorporated herein by reference. (In this regard it is to be noted that Z 5 within the πng structure illustrated above is not to be confused with the moiety Z that, in accordance with the present disclosure, is attached to the NHE-inhibitmg small molecule in order effective render the resulting "NHE-Z" molecule substantially impermeable )
In yet another particular embodiment, the following small molecule,0 disclosed in U S Patent Nos 6,911,453 and 6,703,405, the entire contents of which (and in particular the text of columns 1-7 and 46 of 6,911,453 and columns 14-15 of 6,703,405) are incorporated herein by reference for all relevant and consistent purposes, may be suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is5 substantially impermeable or substantially systemically non-bioavailable)
Figure imgf000051_0002
The vaπables in the structure are defined in the cited patents, the details of which areQ incorporated herein by reference A particularly preferred small molecule falling within the above-noted structure is further illustrated below (see, e g , Example 1 of the 6,911,453 patent, the entire contents of which are specifically incorporated herein by reference)
Figure imgf000052_0001
In yet another particular embodiment, the following small molecules, disclosed in U S Patent Publication Nos 2004/0039001, 2004/0224965, 2005/0113396 and 2005/0020612, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes, may be suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
Figure imgf000052_0002
The vaπables in the structures are defined above and/or m one or more of the cited patent applications, the details of which are incorporated herein by reference, and/or as illustrated above (wherein the broken bonds indicate a point of attachment for the Y moiety to the fused heterocyclic πng) In particular, in vaπous embodiments the combination of X and Y may be as follows
NR6
X = Ar and Y= J^5*6 or ^X
NR7R8
-ξ-N NR7R8
R5
(see, e g , US 2004/0039001 , p 1 therein)
p 1 therein)
Figure imgf000053_0001
[see, e g , US 2005/0113396, p 1 therein)
X = Het and Y = NH2 or NH
-(NH)2-N'' NHR5 -(NH)2-N^NHR5 H
Figure imgf000053_0002
(see, e g , US 2005/00020612, p 1 therein)
In a particularly preferred embodiment of the above-noted structure, the small molecule has the general structure
Figure imgf000053_0003
wherein Ri, R2 and R3 may be the same or different, but are preferably different, and are independently selected from H, NR'R" (wherein R' and R" are independently selected from H and hydrocarbyl, such as lower alkyl, as defined elsewhere herein) and the structure )
In a more particularly preferred embodiment of the above structure, a small molecule falling withm the above-noted structure is further illustrated below (see, e g , compound Il on p 5 of the 2005/0020612 patent application, the entire contents of which are specifically incorporated herein by reference)
Figure imgf000054_0001
In another particularly preferred embodiment, the following small molecule, disclosed in U S Patent No 6,399,824, the entire content of which (and in particular the text of Example 1 therein) is incorporated herein by reference for all relevant and consistent purposes, may be particularly suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
Figure imgf000054_0002
In the structure, R may be preferably selected from H and (CHa)2NCH2CH2-, with H being particularly preferred in various embodiments In yet another particular embodiment, the following small molecule, disclosed in U S Patent No 6,005,010 (and in particular columns 1-3 therein), and/or U S Patent No 6,166,002 (and in particular columns 1-3 therein), the entire contents of which are incorporated herein by reference for all relevant and consistent purposes, may be suitable for use or modification m accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
Figure imgf000055_0001
The vaπable ("R") in the structure is defined in the cited patent application, the details of which are incorporated herein by reference
In yet another particularly preferred embodiment, the following small molecule, disclosed in U S Patent Application No 2008/0194621, the entire content of which (and in particular the text of Example 1 therein) is incorporated herein by reference for all relevant and consistent purposes, may be particularly suitable for use or modification m accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
Ri R2 RJ
Figure imgf000056_0001
-H -NH2 -H
-H -NH2
The vanables ("Ri", "R2 and "R3") in the structure are as defined above, and/or as defined m the cited patent application, the details of which are incorporated herein by reference
In yet another particularly preferred embodiment, the following small molecule, disclosed in U S Patent Application No 2007/0225323, the entire content of which (and in particular the text of Example 36 therein) is incorporated herein by reference for all relevant and consistent purposes, may be particularly suitable for use or
10 modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
Figure imgf000056_0002
I5
In yet another particularly preferred embodiment, the following small molecule, disclosed in U S Patent No 6,911,453, the entire content of which (and m particular the text of Example 35 therein) is incorporated herein by reference for all relevant and consistent purposes, may be particularly suitable for use or modification in accordance with the present disclosure (e g , bound to or modified to include Z, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non-bioavailable)
Figure imgf000057_0001
In one particularly preferred embodiment of the present disclosure, the small molecule may be selected from the group consisting of
Figure imgf000057_0002
In these structures, a bond or link (not shown) may extend, for example, between the Core and amme-substituted aromatic πng (first structure), the heterocyclic πng or the aromatic πng to which it is bound, or alternatively the chloro-substituted aromatic nng (second structure), or the difluoro-substituted aromatic πng or the sulfonamide- substituted aromatic πng (third structure) C. Exemplary Small Molecule Selectivity
Shown below are examples of various NHB inhibiting small molecules and their selectivity across the NHE-I, -2 and -3 isoforms (See, e g , B Masereel et al , An Overview of Inhibitors of Na+ / H+ Exchanger, European J of Med Chem , 38, pp 547-554 (2003), the entire contents of which is incorporated by reference here for all relevant and consistent purposes) Most of these small molecules were optimized as NHE-I inhibitors, and this is reflected in their selectivity with respect thereto (IC50's for subtype- 1 are significantly more potent (numerically lower) than for subtype-3) However, the data in Table 1 indicates that NHE-3 activity may be engineered into an inhibitor seπes originally optimized against a different isoform For example, amiloπde is a poor NHE-3 inhibitor and was inactive against this antiporter at the highest concentration tested (IC50 >100 μM), however, analogs of this compound, such as DMA and EIPA, have NHE-3 IC50's of 14 and 24 uM, respectively The cinnamoylguamdine S-2120 is over 500-fold more active against NHE-I than NHE-3, however, this selectivity is reversed in regioisomer S-3226 It is thus possible to engineer NHE-3 selectivity into a chemical seπes optimized for potency against another antiporter isoform, that is, the inhibitor classes exemplified m the art may be suitably modified for activity and selectivity against NHE-3 (or alternatively NHE-2 and/or NHE-8), as well as being modified to be rendered substantially impermeable or substantially systemically non-bioavailable
Figure imgf000059_0001
Figure imgf000059_0002
Zonipoπde
Figure imgf000059_0004
Figure imgf000059_0003
Figure imgf000059_0005
S-2120 Amiloπde -H -H
DMA -CH3 -CH3
EIPA -C2H5 -CH(CH3J2 HMA -(CH2)6-
Table 1
Figure imgf000059_0006
Figure imgf000060_0001
* - from rat, ** = from rabbit NA - not active a Table adapted from Masereel, B et al , European Journal of Medicinal Chemistry, 2003, 38, 547-54 b K1 values are in italic
As previously noted above, the NHE inhibitor small molecules disclosed herein, including those noted above, may advantageously be modified to render them substantially impermeable or substantially systemically non-bioavailable The compounds as descnbed herein are, accordingly, effectively localized in the gastrointestinal tract or lumen, and in one particular embodiment the colon Since the various NHE isomforms may be found in many different internal organs (e g , bram, heart, liver, etc ), localization of the NHE inhibitors in the intestinal lumen is desirable in order to minimize or eliminate systemic effects (i e , prevent or significantly limit exposure of such organs to these compounds) Accordingly, the present disclosure provides NHE inhibitors, and in particular NHE-3, -2 and/or -8 inhibitors, that are substantially systemically non-bioavailable in the GI tract, and more specifically substantially systemically impermeable to the gut epithelium, as further descnbed below
D. Preferred Embodiments
In one or more particularly preferred embodiments of the present disclosure, the "NHE-Z" molecule is monovalent, that is, the molecule contains one moiety that effectively acts to inhibit NHE-mediated antiport of sodium ions and hydrogen ions In such embodiments, the NHE-Z molecule may be selected, for example, from one of the following structures of Formulas (IV), (V), (VI) or (VII)
Figure imgf000061_0001
wherein each Ri , R2, R3, R5 and R9 are independently selected from H, halogen (e g , Cl), -NR7(CO)R8, -(CO)NR7R8, -SO2-NR7R8, -NR7SO2R8, -NR7R8, -OR7, -SR7, - 0(CO)NR7R8, -NR7(CO)OR8, and -NR7SO2NR8, where R7 and R8 are independently selected from H or Z, where Z is selected from substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, polyalkylene glycol and polyols, where substituents thereon are selected from hydroxyls, amines, amidines, carboxylates, phosphonates, sulfonates, and guamdines, R4 is selected from H, CrC7 alkyl or Z, where Z is selected from substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, a polyalkylene glycol and polyols, where substituents thereon are selected from hydroxyls, amines, amidines, carboxylates, phosphonates, sulfonates, and guamdmes, R6 is absent or selected from H and Ci-C7 alkyl, and, ArI and Ar2 independently represent an aromatic πng, or alternatively a heteroaromatic nng wherein one or more of the carbon atoms therein is replaced with a N, O or S atom,
Figure imgf000061_0002
(V)
wherem each Ri, R2, R3, and R5 are independently selected from H, -NR7(CO)Rs, - (CO)NR7R8, -SO2-NR7R8, -NR7SO2R8, -NR7R8, -OR7, -SR7, -0(CO)NR7R8, - NR7(CO)OR8, and -NR7SO2NR8, where R7 and R8 are independently selected from H or Z, where Z is selected from substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, polyalkylene glycol and polyols, where substituents thereon are selected from hydroxyls, amines, amidines, carboxylates, phosphonates, sulfonates, and guamdines, optionally linked to the πng ArI by a heterocyclic linker, R4 and R12 are independently selected from H and R7, where R7 is as defined above, Rio and Rn, when presented, are independently selected from H and Cj-C7 alkyl, and, ArI and Ar2 independently represent an aromatic πng, or alternatively a heteroaromatic πng wherein one or more of the carbon atoms therein is replaced with a N, O or S atom,
Figure imgf000062_0001
(VII)
I5 wherein each X is a halogen atom, which may be the same or different, Ri is selected from -SO2-NR7R8, -NR7(CO)R8, -(CO)NR7R8, -NR7SO2R8, -NR7R8, -OR7, -SR7, - 0(CO)NR7R8, -NR7(CO)OR8, and -NR7SO2NR8, where R7 and R8 are independently selected from H or Z, where Z is selected from substituted or unsubstituted hydrocarbyl,
20 heterohydrocarbyl, polyalkylene glycol and polyols, where substituents thereon are selected from hydroxyls, amines, amidmes, carboxylates, phosphonates, sulfonates, and guamdines, R3 is selected from H or R7, where R7 is as descπbed above, R13 is selected from substituted or unsubstituted CpCg alkyl, R2 and R]2 are independently selected from H or R7, wherein R7 is as descπbed above, Rio and Ru, when present, are
25 independently selected from H and C]-C7 alkyl, ArI represents an aromatic nng, or alternatively a heteroaromatic πng wherein one or more of the carbon atoms therein is replaced with a N, O or S atom, and Ar2 represents an aromatic πng, or alternatively a heteroaromatic πng wherein one or more of the carbon atoms therein is replaced with a N, O or S atom
In one particular embodiment for the structure of Formula (V), one of Ri, R2 and R3 is linked to the πng ArI, and/or Rs is linked to the πng Ar2, by a 5 heterocyclic linker having the structure
R
O
wherein R represents Rj, R2, R3, or R5 bound thereto
10 In another particular embodiment, the NHE Z molecule of the present disclosure may have the structure of Formula (IV)
Figure imgf000063_0001
I5 wherein each Ri, R2, R3, R5 and R9 are independently selected from H, halogen, NR7(CO)R8, -(CO)NR7R8, -SO2-NR7R8, -NR7SO2R8, -NR7R8, -OR7, -SR7, - 0(CO)NR7R8, -NR7(CO)OR8, and -NR7SO2NR8, where R7 and R8 are independently selected from H or Z, where Z is selected from substituted hydrocarbyl,
20 heterohydrocarbyl, or polyols and/or substituted or unsubstituted polyalkylene glycol, wherein substituents thereon are selected from the group consisting of phosphmates, phosphonates, phosphonamidates, phosphates, phosphonthioates and phosphonodithioates, R4 is selected from H or Z, where Z is substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, a polyalkylene glycol and a polyol, where substituents
25 thereon are selected from hydroxyls, amines, amidines, carboxylates, phosphonates, sulfonates, and guamdines, R^ is selected from -H and C1-C7 alkyl, and, ArI and Ar2 independently represent an aromatic nng, or alternatively a heteroaromatic πng wherein one or more of the carbon atoms therein is replaced with a N, O or S atom.
Additionally, or alternatively, in one or more embodiments of the compounds illustrated above, the compound may optionally have a tPSA of at least about 100 A2, about 150 A2, about 200 A2, about 250 A2, about 270 A2, or more and/or a molecular weight of at least about 710 Da
II. Polyvalent Structures: Macromolccules and Oligomers A. General Structure
As noted above, the compounds of the present disclosure comprise a NHE-mhibitmg small molecule that has been modified or functionahzed structurally to alter its physicochemical properties (by the attachment or inclusion of moiety Z), and more specifically the physicochemical properties of the NHE-Z molecule, thus rendeπng it substantially impermeable or substantially systemically non-bioavailable In one particular embodiment, and as further detailed elsewhere herein, the NHE-Z compound may be polyvalent (1 e , an oligomer, dendrimer or polymer moiety), wherein Z may be referred to in this embodiment generally as a "Core" moiety, and the NHE- inhibiting small molecule may be bound, directly or indirectly (by means of a linking moiety) thereto, the polyvalent compounds having for example one of the following general structures of Formula (VIII), (IX) and (X)
NHE — Core (VIII)
NHE fr^ — Z
J E
(IX)
Core-f — L- -NHE] (X) wherein Core (or Z) and NHE are as defined above, L is a bond or linker, as further defined elsewhere herein below, and E and n are both an integer of 2 or more In vanous alternative embodiments, however, the NHE-inhibiting small molecule may be rendered substantially impermeable or substantially systemically non-bioavailable by forming a polymeric structure from multiple NHE-mhibiting small molecules, which may be the same or different, connected or bound by a seπes of linkers, L, which also may be the same or different, the compound having for example the structure of Formula (XI)
NHE-4— L NHE-] — L NHE
(XI)
wherein Core (or Z) and NHE are as defined above, L is a bond or linker, as further defined elsewhere herein below, and m is 0 or an integer of 1 or more In this embodiment, the physicochemical properties, and in particular the molecular weight or polar surface area, of the NHE-mhibitmg small molecule is modified (e g , increased) by having a seπes of NHE-mhibitmg small molecules linked together, m order to render them substantially impermeable or substantially systemically non-bioavailable In these or yet additional alternative embodiments, the polyvalent compound may be m dimeπc, oligomenc or polymeric form, wherein for example Z or the Core is a backbone to which is bound (by means of a linker, for example) multiple NHE-inhibitmg small molecules Such compounds may have, for example, the structures of Formulas (XIIA) or (XIIB)
— I repeat unrt-j— j L NHE
\ / n
(XIIA) ( — [ repeat unit-| — J
\ I / n
(XIIB)
wherein L is a linking moiety, NHE is a NHE-inhibitmg small molecule, each NHE as descπbed above and in farther detail hereinafter, and n is a non-zero integer (i e , an integer of 1 or more)
The Core moiety has one or more attachment sites to which NHE- mhibiting small molecules are bound, and preferably covalently bound, via a bond or linker, L The Core moiety may, m general, be anything that serves to enable the overall compound to be substantially impermeable or substantially systemically non- bioavailable (e g , an atom, a small molecule, etc ), but in one or more preferred embodiments is an oligomer, a dendπmer or a polymer moiety, in each case having more than one site of attachment for L (and thus for the NHE inhibiting small molecule) The combination of the Core and NHE-mhibitmg small molecule (i e , the "NHE-Z" molecule) may have physicochemical properties that enable the overall compound to be substantially impermeable or substantially systemically non- bioavailable
In this regard it is to be noted that the repeat unit in Formulas (XIIA) and (XIIB) generally encompasses repeating units of vaπous polymeric embodiments, which may optionally be produced by methods referred to herein In each polymeric, or more general polyvalent, embodiment, it is to be noted that each repeat unit may be the same or different, and may or may not be linked to the NHE-inhibiting small molecule by a linker, which in rum may be the same or different when present In this regard it is to be noted that as used herein, "polyvalent" refers to a molecule that has multiple (e g , 2, 4, 6, 8, 10 or more) NHE-inhibitmg moieties therein
In this regard it is to be still farther noted that, as further illustrated elsewhere herein, certain polyvalent NHE-inhibiting compounds of the present disclosure show unexpectedly higher potency, as measured by inhibition assays (as farther detailed elsewhere herein) and charactenzed by the concentration of said NHE inhibitor resulting in 50% inhibition (l e , the IC50 values) It has been observed that certain multivalent structures, represented generally by Formula (X), above, have an IC50 value several fold lower in magnitude than the individual NHE, or L-NHE, structure (which may be referred to as the "monomer" or monovalent form) For example, in one embodiment, multivalent compounds according to Formula (X) were observed to have an IC50 value of at least about 5 time lower (1 e potency about 5 time higher) than the monomer (or monovalent) form (e g Examples 46 and 49) In another embodiment, multivalent compounds according to Formula (X) were observed to have an IC50 value of at least about 10 time lower (1 e potency about 10 time higher) than the monomer form (e g Examples 87 and 88)
The above noted embodiments are further illustrated herein below For example, the first representation below of an exemplary oligomer compound, wherein the various parts of the compound corresponding to the structure of Formula (X) are identified, is intended to provide a broad context for the disclosure provided herein It is to be noted that while each "NHE" moiety (1 e , the NHE small molecule) in the structure below is the same, it is within the scope of this disclosure that each is independently selected and may be the same or different In the illustration below, the linker moiety is a polyethylene glycol (PEG) motif PEG deπvatives are advantageous due m part to their aqueous solubility, which may help avoid hydrophobic collapse (the intramolecular interaction of hydrophobic motifs that can occur when a hydrophobic molecule is exposed to an aqueous environment (see, e g , Wiley, R A , Rich, D H Medicai Research Reviews 1993, 13(3), 327-384) The core moiety illustrated below is also advantageous because it provides some rigidity to the Core — (L — NHE)n molecule, allowing an increase m distance between the NHE inhibitors while minimally increasing rotational degrees of freedom
"Core"
Figure imgf000068_0001
In an alternative embodiment (e g , Formula (XI), wherein m = 0), the structure may be for example
Figure imgf000068_0002
Within the polyvalent compounds utilized for treatments according to the present disclosure, n and m (when m is not zero) may be independently selected from the range of from about 1 to about 10, more preferably from about 1 to about 5, and even more preferably from about 1 to about 2 In alternative embodiments, however, n and m may be independently selected from the range of from about 1 to about 500, preferably from about 1 to about 300, more preferably from about 1 to about 100, and most preferably from about 1 to about 50 In these or other particular embodiments, n and m may both be withm the range of from about 1 to about 50, or from about 1 to about 20
The structures provided above are illustrations of one embodiment of compounds utilized for administration wherein absorption is limited (i e , the compound is rendered substantially impermeable or substantially systemically non-bioavailable) by means of increasing the molecular weight of the NHE-inhibitmg small molecule In an alternative approach, as noted elsewhere herein, the NHE-inhibitmg small molecule may be rendered substantially impermeable or substantially systemically non- bioavailable by means of alteπng, and more specifically increasing, the topological polar surface area, as further illustrated by the following structures, wherein a substituted aromatic rmg is bound to the "scaffold" of the NHE-inhibition small molecule The selection of lomzable groups such as phosphonates, sulfonates, guamdmes and the like may be particularly advantageous at preventing paracellular permeability Carbohydates are also advantageous, and though uncharged, significantly increase tPSA while minimally increasing molecular weight
Figure imgf000069_0001
PSA-alterning moiety It is to be noted, within one or more of the vaπous embodiments illustrated herein, NHE-mhibiting small molecules suitable for use (i e , suitable for modification or functionalization, in order to render them substantially impermeable or substantially systemically non-bioavailable) may, m particular, be selected independently from one or more of the small molecules descπbed as benzoylguandines, heteroaroylguandines, "spacer-stretched" aroylguandmes, non-acyl guamdines and acylguanidine isosteres, above, and as discussed in further detail hereinafter and/or to the small molecules detailed in, for example US5866610, US6399824, US6911453, US6703405, US6005010, US6887870, US6737423, US7326705, US 55824691 (WO94/026709), US6399824 (WO02/024637), US 2004/0339001 (WO02/020496), US 2005/0020612 (WO03/055490), WO01/072742, CA 2387529 (WO01021582), CA 02241531 (WO97/024113), US 2005/0113396 (WO03/051866), US2005/0020612, US2005/0054705, US2008/0194621, US2007/0225323, US2004/0039001, US2004/0224965, US2005/0113396, US2007/0135383, US2007/0135385, US2005/0244367, US2007/0270414, and CA 2177007 (EP0744397), the entire contents of which are incorporated herein by reference for all relevant and consistent purposes Again, it is to be noted that when it is said that NHE-mhibiting small molecule is selected independently, it is intended that, for example, the ohgomeπc structures represented m Formulas (X) and (XI) above can include different structures of the NHE small molecules, within the same oligomer or polymer In other words, each "NHE" within a given polyvalent embodiment may independently be the same or different than other "NHE" moieties withm the same polyvalent embodiment
In designing and making the substantially impermeable or substantially systemically non-bioavailable, NHE-mhibiting compounds that may be utilized for the treatments detailed in the instant disclosure, it may in some cases be advantageous to first determine a likely point of attachment on a small molecule NHE inhibitor, where a core or linker might be installed or attached before making a senes of candidate multivalent or polyvalent compounds This may be done by one skilled in the art via known methods by systematically installing functional groups, or functional groups displaying a fragment of the desired core or linker, onto vaπous positions of the NHE inhibitor small molecule and then testing these adducts to determine whether the modified inhibitor still retains desired biological properties (e g , NHE inhibition) An understanding of the SAR of the inhibitor also allows the design of cores and/or linkers that contribute positively to the activity of the resulting compounds For example, the SAR of an NHE inhibitor seπes may show that installation of an N-alkylated piperazme contributes positively to biochemical activity (increased potency) or pharmaceutical properties (increased solubility), the piperazme moiety may then be utilized as the point of attachment for the desired core or linker via N-alkylation In this fashion, the resulting compound thereby retains the favorable biochemical or pharmaceutical properties of the parent small molecule In another example, the SAR of an NHE inhibitor series might indicate that a hydrogen bond donor is important for activity or selectivity Core or linker moieties may then be designed to ensure this H-bond donor is retained These cores and/or linkers may be further designed to attenuate or potentiate the pKa of the H-bond donor, potentially allowing improvements in potency and selectivity In another scenaπo, an aromatic πng in an inhibitor could be an important pharmacophore, interacting with the biological target via a pi-stackmg effect or pi-cation interaction Linker and core motifs may be similarly designed to be isostenc or otherwise synergize with the aromatic features of the small molecule Accordingly, once the structure- activity relationships within a molecular seπes are understood, the molecules of interest can be broken down into key pharmacophores which act as essential molecular recognition elements When consideπng the installation of a core or linker motif, said motifs can be designed to exploit this SAR and may be installed to be isostenc and isoelectronic with these motifs, resulting in compounds that retain biological activity but have significantly reduced permeability
Another way the SAR of an inhibitor seπes can be exploited in the installation of core or linker groups is to understand which regions of the molecule are insensitive to structural changes For example, X-ray co-crystal structures of protein- bound inhibitors can reveal those portions of the inhibitor that are solvent exposed and not involved in productive interactions with the target Such regions can also be identified empiπcally when chemical modifications in these regions result in a "flat SAR" (i e , modifications appear to have minimal contribution to biochemical activity) Those skilled in the art have frequently exploited such regions to engineer in pharmaceutical properties into a compound, for example, by installing motifs that may improve solubility or potentiate ADME properties In the same fashion, such regions are expected to be advantageous places to install core or linker groups to create compounds as descπbed in the instant disclosure These regions are also expected to be sites for adding, for example, highly polar functionality such as carboxylic acids, phosphomc acids, sulfonic acids, and the like in order to greatly increase tPSA
Another aspect to be considered in the design of cores and linkers displaying an NHE inhibitor is the limiting or preventing of hydrophobic collapse Compounds with extended hydrocarbon functionalities may collapse upon themselves in an intramolecular fashion, causing an increased enthalpic barrier for interaction with the desired biological target Accordingly, when designing cores and linkers, these are preferably designed to be resistant to hydrophobic collapse For example, conformational constraints such as πgid monocyclic, bicyclic or polycychc πngs can be installed in a core or linker to increase the rigidity of the structure Unsaturated bonds, such as alkenes and alkynes, may also or alternatively be installed Such modifications may ensure the NHE-inhibiting compound is accessible for productive binding with its target Furthermore, the hydrophilicity of the linkers may be improved by adding hydrogen bond donor or acceptor motifs, or ionic motifs such as amines that are protonated in the GI, or acids that are deprotonated Such modifications will increase the hydrophilicity of the core or linker and help prevent hydrophobic collapse Furthermore, such modifications will also contribute to the impermeability of the resulting compounds by increasing tPSA
Specific examples of NHE-inhibitmg small molecules modified consistent with the principles detailed above are illustrated below These moieties display functional groups that facilitate their appendage to "Z" (e g , a core group, Core, or linking group, L) These functional groups can include electrophiles, which can react with nucleophilic cores or linkers, and nucleophiles, which can react with electrophilic cores or linkers Small molecule NHE inhibitors may be similarly deπvatized with, for example, boronic acid groups which can then react with appropπate cores or linkers via palladium mediated cross-coupling reactions The NHE inhibitor may also contain olefins which can then react with appropπate cores or linkers via olefin metathesis chemistry, or alkynes or azides which can then react with appropπate cores or linkers via [2 + 3] cycloaddtion One skilled in the art may consider a vaπety of functional groups that will allow the facile and specific attachment of an NHE inhibiting small molecule to a desired core or linker Exemplary functioπahzed derivatives of NHEs include but are not limited to the following
Scheme 1
Cinnamoylguamdine NHE-inhibitmg Moiety Functionahzed to Display Electrophilic or Nucleophilic Groups to Facilitate Reaction with Cores and Linkers
Figure imgf000073_0001
Electrophilic Intermediates Nucleophilic Intermediates „
«
Figure imgf000073_0002
wherein the variables in the above-noted structures (e g , R, etc ) are as defined in U S Patent No 6,399,824, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes Scheme 2
Tetrahydroisoqumohne NHE-inhibiting Moiety Functionabzed to Display Electrophilic or Nucleophilic Groups to Facilitate Reaction with Cores and Linkers
Figure imgf000074_0001
Nucleophilic Intermediates Electrophilic Intermediates
Figure imgf000074_0002
wherein the vaπables in the above-noted structures (e g , Rγ 9, etc ) are as defined m U S Patent No 6,911,453, the entire contents of which (and in particular the text of columns 1-4 therein) are incorporated herein by reference for all relevant and consistent purposes Scheme 3
Qmnazolme NHE-inhibitmg Moiety Functionahzed to Display Electrophilic or Nucleophilic Groups to Facilitate Reaction with Cores and Linkers
Figure imgf000075_0001
Nucleophilic Intermediates. Electrophilic Intermediates
Figure imgf000075_0002
wherein the vaπables in the above-noted structures (e g , R7 9, etc ) are as defined m U S Patent Application No 2005/0020612 and U S Patent No 6,911,453, the entire contents of which (and in particular the text of columns 1-4 therein) are incorporated herein by reference for all relevant and consistent purposes
It is to be noted that one skilled in the art can envision a number of core or linker moieties that may be functionahzed with an appropnate electrophile or nucleophile Shown below are a seπes of such compounds selected based on several design considerations, including solubility, steric effects, and their ability to confer, or be consistent with, favorable structure-activity relationships. In this regard it is to be further noted, however, that the structures provided below, and above, are for illustration purposes only, and therefore should not be viewed in a limiting sense.
Exemplary electrophilic and nucleophilic linker moieties include, but are not limited to, the linker moieties illustrated in the Examples and the following:
Nucleophilic linkers (for use with electrophilic NHE-inhibitory derivatives)
etc ,
Figure imgf000076_0001
kDa, etc
-CHO, -OH, -SH, etc
Figure imgf000076_0002
Electrophilic linkers (for use with nucleophilic NHE-inhibitory derivatives)
Figure imgf000076_0003
n = * 0-,v 1, 2, 3, 4*, etc n = 1 , 2, 3, 4, etc π = 2, 3, 4, etc., X = -OH, -Cl, -NHS, etc X = -OH, -Cl, -NHS, etc 3.4 kDa, 5 kDa, etc. R = tosyl, mesyl, etc
0HC^,0^0^CHO n XO2C n = 2, 3, 4, etc., 34 kDa, 5 kDa, etc π = 2, " 3X, 4, 5, 6, e6tc n = 1, 2, 3, etc R = tosyl, mesyl, etc X = -Cl, -Br, -OTs, etc X = -Cl, -NHS, OH, etc
N^CO2X
XO^^N^J ^Or-R n = 2, 3, 4, etc , n = 1, 2, 3, etc 34 kDa, 5 kDa, etc
X = -Cl, -NHS, OH, etc Ri = tosyl, mesyl, etc
R2 = -N3, -CO2H, -CHO, -OH, -SH,
-C=CH2, -C=CH, etc
The linking moiety, L, in each of the described embodiments (including embodiments in which a NHE-inhibiting small molecule is linked to a core such as an atom, another small molecule, a polymer moiety, an oligomer moiety, or a nonrepeating moiety) can be a chemical linker, such as a bond or other moiety, for example, comprising about 1 to about 200 atoms, or about 1 to about 100 atoms, or about 1 to about 50 atoms, that can be hydrophilic and/or hydrophobic In one embodiment, the linking moiety can be a polymer moiety grafted onto a polymer backbone, for example, using living free radical polymerization approaches known in the art Preferred L structures or moieties may also be selected from, for example, ohgoethylene glycol, oligopeptide, oligoethyleneimme, ohgotetramethylene glycol and oligocaprolactone As noted, the core moiety can be an atom, a small molecule, an oligomer, a dendπmer or a polymer moiety, in each case having one or more sites of attachment for L For example, the core moiety can be a non-repeating moiety (considered as a whole including linking points to the inhibitors), selected for example from the group consisting of alkyl, phenyl, aryl, alkenyl, alkynyl, heterocyclic, amine, ether, sulfide, disulfide, hydrazine, and any of the foregoing substituted with oxygen, sulfur, sulfonyl, phosphonyl, hydroxyl, alkoxyl, amine, thiol, ether, carbonyl, carboxyl, ester, amide, alkyl, alkenyl, alkynyl, aryl, heterocyclic, and moieties comprising combinations thereof (in each permutation) A non-repeating moiety can include repeating units (e g , methylene) within portions or segments thereof (e g , within an alkyl segment), without having discrete repeat units that constitute the moiety as a whole (e g , in the sense of a polymer or oligomer)
Exemplary core moieties include but are not limited to the core moieties illustrated in the Examples and ether moieties, ester moieties, sulfide moieties, disulfide moieties, amine moieties, aryl moieties, alkoxyl moieties, etc , such as, for example, the following
Figure imgf000078_0001
Figure imgf000078_0002
Figure imgf000079_0001
wherein the broken bonds (i e , those having a wavy bond, ^ , through them) are points of connection to either an NHE inhibitor or a linker moiety displaying an NHE inhibitor, where said points of connection can be made using chemistπes and functional groups known to the art of medicinal chemistry, and further wherein each p, q, r and s is an independently selected integer ranging from about 0 to about 48, preferably from about 0 to about 36, or from about 0 to about 24, or from about 0 to about 16. In some instances, each p, q, r and s can be an independently selected integer ranging from about 0 to 12. Additionally, R can be a substituent moiety generally selected from halide, hydroxyl, amine, thiol, ether, carbonyl, carboxyl, ester, amide, carbocyclic, heterocyclic, and moieties comprising combinations thereof.
In another approach, the core moiety is a dendrimer, defined as a repeatedly branched molecule (see, e.g., J. M. J. Frechet, D. A. Tomalia, Dendrimers and Other Dendritic Polymers, John Wiley & Sons, Ltd. NY, NY, 2001) and schematically represented below:
Figure imgf000080_0001
DENDRIMER DENORON
In this approach, the NHE inhibiting small molecule is attached through L to one, several or optionally all termini located at the periphery of the dendrimer. In another approach, a dendπmer building block named dendron, and illustrated above, is used as a core, wherein the NHE inhibitor group is attached to one, several or optionally all termini located at the periphery of the dendron. The number of generations herein is typically between about 0 and about 6, and preferably between about 0 and about 3. (Generation is defined in, for example, J. M. J. Frechet, D. A. Tomalia, Dendrimers and Other Dendritic Polymers, John Wiley & Sons, Ltd. NY, NY.) Dendrimer and/or dendron structures are well known in the art and include, for example, those shown in or illustrated by: (i) J. M. J. Frechet, D. A. Tomalia, Dendrimers and Other Dendritic Polymers, John Wiley & Sons, Ltd NY, NY, (π) George R Newkome, Charles N Moorefield and Fπtz Vogtle, Dendnmers and Dendrons Concepts, Syntheses, Applications, VCH Verlagsgesellschaft Mbh, and, (in) Boas, U , Chπstensen, J B , Heegaard, P M H , Dendnmers in Medicine and Biotechnology New Molecular Tools , Springer, 2006
In yet another approach, the core moiety may be a polymer moiety or an oligomer moiety The polymer or oligomer may, in each case, be independently considered and compose repeat units consisting of a repeat moiety selected from alkyl {e g , -CH2-), substituted alkyl (e g , -CHR- , wherein, for example, R is hydroxy), alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, phenyl, aryl, heterocyclic, amine, ether, sulfide, disulfide, hydrazine, and any of the foregoing substituted with oxygen, sulfur, sulfonyl, phosphonyl, hydroxyl, alkoxyl, amine, thiol, ether, carbonyl, carboxyl, ester, amide, alkyl, alkenyl, alkynyl, aryl, heterocyclic, as well as moieties compπsing combinations thereof In still another approach, the core moiety compπses repeat units resulting from the polymerization of ethylenic monomers (e g , such as those ethylenic monomers listed elsewhere herein below)
Preferred polymers for polymeric moieties useful in constructing substantially impermeable or substantially systemically non-bioavailable NHE- inhibitmg compounds that are multivalent, for use in the treatment vaπous treatment methods disclosed herein, can be prepared by any suitable technique, such as by free radical polymerization, condensation polymerization, addition polymerization, ring- opening polymerization, and/or can be derived from naturally occurring polymers, such as saccharide polymers Further, in some embodiments, any of these polymer moieties may be functionahzed Examples of polysacchaπdes useful in preparation of such compounds include but are not limited to mateπals from vegetable or animal origin, including cellulose mateπals, hemicellulose, alkyl cellulose, hydroxyalkyl cellulose, carboxymethylcellulose, sulfoethylcellulose, starch, xylan, amylopectine, chondroitm, hyarulonate, heparin, guar, xanthan, mannan, galactomannan, chitin, and/or chitosan More preferred, in at least some instances, are polymer moieties that do not degrade, or that do not degrade significantly, under the physiological conditions of the GI tract (such as, for example, carboxymethylcellulose, chitosan, and sulfoethylcellulose)
When free radical polymerization is used, the polymer moiety can be prepared from vaπous classes of monomers including, for example, acrylic, methacrylic, styremc, vinylic, and dienic, whose typical examples are given thereafter styrene, substituted styrene, alkyl acrylate, substituted alkyl acrylate, alkyl methacrylate, substituted alkyl methacrylate, acrylomtπle, methacrylomtnle, acrylamide, methacrylamide, N-alkylacrylamide, N-alkylmethacrylamide, N,N- dialkylacrylamide, N,N-dialkylmethacrylamide, isoprene, butadiene, ethylene, vmyl acetate, and combinations thereof Functionahzed versions of these monomers may also be used and any of these monomers may be used with other monomers as comonomers For example, specific monomers or comonomers that may be used in this disclosure include methyl methacrylate, ethyl methacrylate, propyl methacrylate (all isomers), butyl methacrylate (all isomers), 2-ethylhexyl methacrylate, isobomyl methacrylate, methacrylic acid, benzyl methacrylate, phenyl methacrylate, methacrylomtnle, α-methylstyrene, methyl acrylate, ethyl acrylate, propyl acrylate (all isomers), butyl acrylate (all isomers), 2-ethylhexyl acrylate, isobomyl acrylate, acrylic acid, benzyl acrylate, phenyl acrylate, acrylomtπle, styrene, glycidyl methacrylate, 2- hydroxyethyl methacrylate, hydroxypropyl methacrylate (all isomers), hydroxybutyl methacrylate (all isomers), N,N-dimethylammoethy] methacrylate, N,N- diethylammoethyl methacrylate, tπethyleneglycol methacrylate, itacomc anhydride, itacomc acid, glycidyl acrylate, 2-hydroxyethyl acrylate, hydroxypropyl acrylate (all isomers), hydroxybutyl acrylate (all isomers), N,N-dimethylaminoethyl acrylate, N,N- diethylaminoethyl acrylate, tπethyleneglycol acrylate, methacrylamide, N- methylacrylamide, N,N-dimethylacrylamide, N-tert-butylmethacrylamide, N-n- butylmethacryl amide, N-methylolmethacryl amide, N-ethylolmethacrylamide, N-tert- butylacrylamide, N-N-butylacrylamide, N-methylolacrylamide, N-ethylolacrylamide, 4- acryloylmoφhohne, vmyl benzoic acid (all isomers), diethylammostyrene (all isomers), a-methylvmyl benzoic acid (all isomers), diethylammo α-methylstyrene (all isomers), p- vinylbenzene sulfonic acid, p-vmylbenzene sulfonic sodium salt, alkoxy and alkyl silane functional monomers, maleic anhydπde, N-phenylmaleimide, N-butylmaleimide, butadiene, isoprene, chloroprene, ethylene, vinyl acetate, vinylformamide, allylamine, vmylpyπdmes (all isomers), fluoπnated acrylate, methacrylates, and combinations thereof Mam chain heteroatom polymer moieties can also be used, including polyethyleneimine and polyethers such as polyethylene oxide and polypropylene oxide, as well as copolymers thereof
In one particular embodiment, the polymer to which the NHE inhibitor small molecule, NHE, is attached or otherwise a part of is a polyol (e g , a polymer having a repeat unit of, for example, a hydroxyl-substituted alkyl, such as -CH(OH)-) Polyols, such as mono- and disaccharides, with or without reducing or reducible end groups thereon, may be good candidates, for example, for installing additional functionality that could render the compound substantially impermeable
In one particular embodiment, the NHE inhibiting small molecule, NHE, is attached at one or both ends of the polymer chain More specifically, in yet another alternative approach to the polyvalent embodiment of the present disclosure, a macromolecule (e g , a polymer or oligomer) having one of the following exemplary structures may be designed and constructed as descπbed herein
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000084_0002
Figure imgf000085_0001
Figure imgf000086_0001
It is to be further noted that the repeat moiety m Formulas (XIIA) or (XIIB) generally encompasses repeating units of polymers and copolymers produced by methods referred to herein above
It is to be noted that the various properties of the oligomers and polymers that form the core moiety as disclosed herein above may be optimized for a given use or application using experimental means and principles generally known m the art For example, the overall molecular weight of the compounds or structures presented herein above may be selected so as to achieve non-absorbability, inhibition persistence and/or potency
Additionally, with respect to those polymeric embodiments that encompass or include the compounds generally represented by the structure of Formula (I) herein, and/or those disclosed for example in the many patents and patent applications cited herein (see, e g , US5866610, US6399824, US691 1453, US6703405, US6005010, US6887870, US6737423, US7326705, US 55824691 (WO94/026709), US6399824 (WO02/024637), US 2004/0339001 (WO02/020496), US 2005/0020612 (WO03/055490), WO01/072742, CA 2387529 (WO01021582), CA 02241531 (WO97/0241 13), US 2005/01 13396 (WO03/051866), US2005/0020612, US2005/0054705, US2008/0194621 , US2007/0225323, US2004/0039001 , US2004/0224965, US2005/01 13396, US2007/0135383, US2007/0135385, US2005/0244367, US2007/0270414, and CA 2177007 (EP0744397), the entire contents of which are incorporated herein by reference for all relevant and consistent purposes), such as those wherem these compounds or structures are pendants off of a polymeric backbone or chain, the composition of the polymeric backbone or chain, as well as the overall size or molecular weight of the polymer, and/or the number of pendant molecules present thereon, may be selected according to various principles known in the art in view of the intended application or use
With respect to the polymer composition of the NHE inhibiting compound, it is to be noted that a number of polymers can be used including, for example, synthetic and/or naturally occurring aliphatic, ahcychc, and/or aromatic polymers In preferred embodiments, the polymer moiety is stable under physiological conditions of the GI tract By "stable" it is meant that the polymer moiety does not degrade or does not degrade significantly or essentially does not degrade under the physiological conditions of the GI tract For instance, at least about 90%, preferably at least about 95%, and more preferably at least about 98%, and even more preferably at least about 99% of the polymer moiety remains un-degraded or intact after at least about 5 hours, at least about 12 hours, at least about 18 hours, at least about 24 hours, or at least about 48 hours of residence in a gastrointestinal tract Stability in a gastrointestinal tract can be evaluated using gastrointestinal mimics, e g , gastric mimics or intestinal mimics of the small intestine, which approximately model the physiological conditions at one or more locations therein Polymer moieties detailed herein for use as the core moiety can be hydrophobic, hydrophihc, amphiphilic, uncharged or non-ionic, negatively or positively charged, or a combination thereof Additionally, the polymer architecture of the polymer moiety can be linear, grafted, comb, block, star and/or dendπtic, preferably selected to produce desired solubility and/or stability characteristics as descπbed above.
Additionally or alternatively, modifications may be made to NHE-
5 inhibiting small molecules that increase tPSA, thus contributing to the impermeability of the resulting compounds Such modifications preferably include addition of di- anions, such as phosphonates, malonates, sulfonates and the like, and polyols such as carbohydrates and the like Exemplary derivatives of NHEs with increased tPSA include but are not limited to the following
Figure imgf000088_0001
B. Preferred Embodiments
In one or more particularly preferred embodiments of the present disclosure, the "NHE-Z" molecule is polyvalent, that is, the molecule contains two or
15 more moieties that effectively acts to inhibit NHE-mediated antiport of sodium ions and hydrogen ions In such embodiments, the NHE-Z molecule may be selected, for example, from one of the following Formulas (IV), (V), (VI) or (VII)
Figure imgf000089_0001
(IV)
wherein each Ri, R2, R3, R5 and R9 are independently selected from H, halogen, - NR7(CO)R8, -(CO)NR7R8, -SO2-NR7R8, -NR7SO2R8, -NR7R8, -OR7, -SR7, - 0(CO)NR7R8, -NR7(CO)OR8, and -NR7SO2NR8, where R7 and R8 are independently selected from H or L, provided at least one is L, wherein L is selected from the group consisting of substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, polyalkylene glycol and polyols, and further wherein L links the repeat unit to at least one other repeat unit and/or at least one other Core moiety independently selected from substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, polyalkylene glycol, polyols, polyamines, or polyacrylamides, of the polyvalent compound, R4 is selected from H, Ci -C7 alkyl or L, where L is as descnbed above, R6 is absent or selected from H and Cj-C7 alkyl, and, ArI and Ar2 independently represent an aromatic πng, or alternatively a heteroaromatic πng wherein one or more of the carbon atoms therein is replaced with a N, O or S atom,
Figure imgf000089_0002
(V)
wherein each Ri, R2, R3, and R5 are optionally linked to the πng ArI by a heterocyclic linker, and further are independently selected from H, -NR7(CO)R8, -(CO)NR7R8, -SO2- NR7R5, NR7SO2R8, -NR7R8, -OR7, -SR7, -0(CO)NR7R8, -NR7(CO)OR8, and - NR7SO2NR8, where R7 and R8 are independently selected from H or L, provided at least one is L, wherein L is selected from the group consisting of substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, polyalkylene glycol and polyols, and further wherein L links the repeat unit to at least one other repeat unit and/or at least one other Core moiety independently selected from substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, polyalkylene glycol, polyols, polyamines, or polyacrylamides, of the polyvalent compound, R4 and R12 are independently selected from H or L, where L is as defined above, Rio and Rn, when presented, are independently selected from H and Ci- C7 alkyl, and, ArI and Ar2 independently represent an aromatic πng, or alternatively a heteroaromatic nng wherein one or more of the carbon atoms therein is replaced with a N, O or S atom,
Figure imgf000090_0001
(VI) or
Figure imgf000090_0002
* (VII)
wherein each X is a halogen atom, which may be the same or different, Ri is selected from -SO2-NR7R8, -NR7(CO)R8, -(CO)NR7R8, -NR7SO2R8, -NR7R8, -OR7, -SR7, - 0(CO)NR7R8, -NR7(CO)OR8, and -NR7SO2NR8, where R7 and R8 are independently selected from H or L, provided at least one is L, wherein L is selected from the group consisting of substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, polyalkylene glycol and polyols, and further wherein L links the repeat unit to at least one other repeat unit and/or at least one other Core moiety independently selected from substituted or unsubstituted hydrocarbyl, heterohydrocarbyl, polyalkylene glycol, polyols, polyamines, or polyacrylamides, of the polyvalent compound, R3 is selected from H or L, where L is as descπbed above, R13 is selected from substituted or unsubstituted Cj-Cg alkyl, R2 and R12 are independently selected from H or L, wherein L is as descπbed above, R10 and R11, when present, are independently selected from H and C1-C7 alkyl, ArI represents an aromatic πng, or alternatively a heteroaromatic πng wherein one or more of the carbon atoms therein is replaced with a N, O or S atom, and Ar2 represents an aromatic nng, or alternatively a heteroaromatic πng wherein one or more of the carbon atoms therein is replaced with a N, O or S atom
In one particular embodiment for the structure of Formula (V), one of Ri, R2 and R3 is linked to the πng ArI, and/or R5 is linked to the πng Ar2, by a heterocyclic linker having the structure
)
wherein R represents Ri, R2, R3, or R5 bound thereto In one particular embodiment, the NHE-inhibitmg small molecule has the structure of Formula (IV)
Figure imgf000091_0001
(IV) or a stereoisomer, prodrug or pharmaceutically acceptable salt thereof, wherein each Ri, R2, R3, R5 and R9 are independently selected from H, halogen, -NRv(CO)Rs, - (CO)NR7R8, -SO2-NR7R8, -NR7SO2R8, -NR7R8, -OR7, -SR7, -0(CO)NR7R8, - NR7(CO)OR8, and -NR7SO2NR8, where R7 and R8 are independently selected from H or a bond linking the NHE-inhibiting small molecule to L, provided at least one is a bond linking the NHE-inhibitmg small molecule to L, R4 is selected from H, Ci-C7 alkyl, or a bond linking the NHE-inhibitmg small molecule to L, Ke is absent or selected from H and C1-C7 alkyl, and ArI and Ar2 independently represent an aromatic nng or a heteroaromatic nng
In further particular embodiments of the above embodiment, the NHE- mhibiting small molecule has the following structure
Figure imgf000092_0001
or a stereoisomer, prodrug or pharmaceutically acceptable salt thereof, wherein each Ri, R2 and R3 are independently selected from H, halogen, -NR7(CO)R8, -(CO)NR7R8, - SO2-NR7R8, NR7SO2R8, -NR7R8, -OR7, -SR7, -0(CO)NR7R8, -NR7(CO)OR8, and - NR7SO2NR8, where R7 and R8 are independently selected from H or a bond linking the NHE-inhibitmg small molecule to L, provided at least one is a bond linking the NHE- inhibiting small molecule to L
In further particular embodiments of the above embodiment, the NHE- mhibitmg small molecule has one of the following structures
Figure imgf000092_0002
or a stereoisomer, prodrug or pharmaceutically acceptable salt thereof
In further particular embodiments of the above embodiment, L is a polyalkylene glycol linker, such as a polyethylene glycol linker
In further particular embodiments of the above embodiment, n is 2
In further particular embodiments of the above embodiment, the Core has the following structure ξ-X-Y-X— f wherein X is selected from the group consisting of a bond, -O-, -NH-, -S-, Ci 6alkylene, -NHC(O)-, -C(=O)NH-, -NHC(=0)NH-, -SO2NH-, and -NHSO2-, Y is selected from the group consisting of a bond, optionally substituted Ci galkylene, optionally substituted aryl, optionally substituted heteroaryl, a polyethylene glycol linker, -(CH2), 6O(CH2)! 6- and -(CH2)] 6NYi(CH2)i 6-, and Yi is selected from the group consisting of hydrogen, optionally substituted Ci salkyl, optionally substituted aryl or optionally substituted heteroaryl In further particular embodiments of the above embodiment, the Core is selected from the group consisting of
Figure imgf000093_0001
III. Terminology, Physical and Performance Properties A. Terminology
Unless the context requires otherwise, throughout the present specification and claims, the word "comprise" and vaπations thereof, such as, "comprises" and "comprising" are to be construed in an open, inclusive sense, that is as "including, but not limited to"
Reference throughout this specification to "one embodiment" or "an embodiment" means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment of the present invention Thus, the appearances of the phrases "in one embodiment" or "in an embodiment" in various places throughout this specification are not necessarily all referring to the same embodiment Furthermore, the particular features, structures, or characteristics may be combined m any suitable manner m one or more embodiments
"Amino" refers to the -NH2 radical "Cyano" refers to the -CN radical
"Hydroxy" or "hydroxyl" refers to the -OH radical
"Immo" refers to the =NH substituent
"Nitro" refers to the -NO2 radical
"Oxo" refers to the -O substituent "Thioxo" refers to the -S substituent
"Alkyl" refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, which is saturated or unsaturated (; e , contains one or more double and/or triple bonds), having from one to twelve carbon atoms (C1-C12 alkyl), preferably one to eight carbon atoms (Ci-Cg alkyl) or one to six carbon atoms (C1-C6 alkyl), and which is attached to the rest of the molecule by a single bond, e g , methyl, ethyl, rc-propyl, 1-methylethyl (wo-propyl), rc-butyl, n-pentyl,
1,1-dimethylethyl (/-butyl), 3-methylhexyl, 2-methylhexyl, ethenyl, prop-1-enyl, but 1-enyl, pent-1-enyl, penta 1,4 dienyl, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like Unless stated otherwise specifically m the specification, an alkyl group may be optionally substituted
"Alkylene" or "alkylene chain" refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, which is saturated or unsaturated (1 e , contains one or more double and/or triple bonds), and having from one to twelve carbon atoms, e g , methylene, ethylene, propylene, «-butylene, ethenylene, propenylene, «-butenylene, propynylene, rc-butynylene, and the like The alkylene chain is attached to the rest of the molecule through a single or double bond and to the radical group through a single or double bond The points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon or any two carbons within the chain Unless stated otherwise specifically in the specification, an alkylene chain may be optionally substituted "Alkoxy" refers to a radical of the formula -ORa where Ra is an alkyl radical as defined above containing one to twelve carbon atoms Unless stated otherwise specifically in the specification, an alkoxy group may be optionally substituted "Alkylamino" refers to a radical of the formula -NHR3 or -NRaRa where each Ra is, independently, an alkyl radical as defined above containing one to twelve carbon atoms Unless stated otherwise specifically in the specification, an alkylamino group may be optionally substituted
"Thioalkyl" refers to a radical of the formula -SRa where Ra is an alkyl radical as defined above containing one to twelve carbon atoms Unless stated otherwise specifically in the specification, a thioalkyl group may be optionally substituted
"Aryl" refers to a hydrocarbon nng system radical compπsing hydrogen, 6 to 18 carbon atoms and at least one aromatic nng For purposes of this invention, the aryl radical may be a monocyclic, bicyclic, tπ cyclic or tetracyclic nng system, which may include fused or bndged nng systems Aryl radicals include, but are not limited to, aryl radicals denved from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, fluoranthene, fluorene, as indacene, 5-indacene, mdane, mdene, naphthalene, phenalene, phenanthrene, pleiadene, pyrene, and tnphenylene Unless stated otherwise specifically in the specification, the term "aryl" or the prefix "ar-" (such as in "aralkyl") is meant to include aryl radicals that are optionally substituted
"Aralkyl" refers to a radical of the formula -Rb-R0 where Rb is an alkylene chain as defined above and R0 is one or more aryl radicals as defined above, for example benzyl, diphenylmethyl and the like Unless stated otherwise specifically in the specification, an aralkyl group may be optionally substituted
"Cycloalkyl" or "carbocyclic nng" refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, which may include fused or bndged nng systems, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molecule by a single bond Monocyclic radicals include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl Polycyclic radicals include, for example, adamantyl, norbornyl, decalmyl, 7,7-dimethyl-bicyclo[2 2 ljheptanyl, and the like Unless otherwise stated specifically in the specification, a cycloalkyl group may be optionally substituted
"Cycloalkylalkyl" refers to a radical of the formula -RbRd where Ra is an alkylene chain as defined above and Rg is a cycloalkyl radical as defined above Unless stated otherwise specifically in the specification, a cycloalkylalkyl group may be optionally substituted "Fused" refers to any ring structure described herein which is fused to an existing πng structure in the compounds of the invention When the fused πng is a heterocyclyl πng or a heteroaryl πng, any carbon atom on the existing πng structure which becomes part of the fused heterocyclyl πng or the fused heteroaryl πng may be replaced with a nitrogen atom "Halo" or "halogen" refers to bromo, chloro, fluoro or iodo
"Haloalkyl" refers to an alkyl radical, as defined above, that is substituted by one or more halo radicals, as defined above, e g , tπfluoromethyl, difluoromethyl, tnchloromethyl, 2,2,2-tπfluoroethyl, 1 ,2-difluoroethyl,
3-bromo-2-fluoropropyl, 1,2-dibromoethyl, and the like Unless stated otherwise specifically in the specification, a haloalkyl group may be optionally substituted
"Heterocyclyl" or "heterocyclic πng" refers to a stable 3- to 18-membered non-aromatic πng radical which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur Unless stated otherwise specifically in the specification, the heterocyclyl radical may be a monocyclic, bicychc, tricyclic or tetracyclic πng system, which may include fused or bπdged πng systems, and the nitrogen, carbon or sulfur atoms in the heterocyclyl radical may be optionally oxidized, the nitrogen atom may be optionally quaternized, and the heterocyclyl radical may be partially or fully saturated Examples of such heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl[l,3]dithianyl, decahydroisoqumolyl, lmidazohnyl, lmidazolidmyl, lsothiazohdinyl, lsoxazohdinyl, morphohnyl, octahydroindolyl, octahydroisomdolyl, 2-oxopiperazmyl, 2-oxopipeπdmyl, 2-oxopyrrolidinyl, oxazobdmyl, pipeπdmyl, piperazmyl, 4-pipeπdonyl, pyrrohdinyl, pyrazohdinyl, qumuchdmyl, thiazohdmyl, tetrahydrofuryl, tnthianyl, tetrahydropyranyl, thiomorpholmyl, tfiiamorpholinyl, 1-oxo-thiomorpholmyl, and 1,1-dioxo-thiomoφhohnyl Unless stated otherwise specifically m the specification, Unless stated otherwise specifically in the specification, a heterocyclyl group may be optionally substituted
"iV-heterocyclyl" refers to a heterocyclyl radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a nitrogen atom in the heterocyclyl radical Unless stated otherwise specifically in the specification, a iV-heterocyclyl group may be optionally substituted
"Heterocyclylalkyl" refers to a radical of the formula -RbRe where Rb is an alkylene chain as defined above and Re is a heterocyclyl radical as defined above, and if the heterocyclyl is a mtrogen-contaimng heterocyclyl, the heterocyclyl may be attached to the alkyl radical at the nitrogen atom Unless stated otherwise specifically m the specification, a heterocyclylalkyl group may be optionally substituted
"Heteroaryl" refers to a 5- to 14-membered πng system radical compπsing hydrogen atoms, one to thirteen carbon atoms, one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur, and at least one aromatic πng For purposes of this invention, the heteroaryl radical may be a monocyclic, bicychc, tricyclic or tetracyclic πng system, which may include fused or bπdged nng systems, and the nitrogen, carbon or sulfur atoms in the heteroaryl radical may be optionally oxidized, the nitrogen atom may be optionally quatermzed Examples include, but are not limited to, azepinyl, acπdinyl, benzimidazolyl, benzothiazolyl, benzindolyl, benzodioxolyl, benzofuranyl, benzooxazolyl, benzothiazolyl, benzothiadiazolyl, benzo[6][l,4]dioxepinyl, 1 ,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxmyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzotπazolyl, benzo[4,6]imidazo[l,2-a]pyπdinyl, carbazolyl, cinnolmyl, dibenzofuranyl, dibenzothiophenyl, furanyl, furanonyl, isothiazolyl, lmidazolyl, indazolyl, indolyl, mdazolyl, lsomdolyl, mdohnyl, lsomdolmyl, isoquinolyl, mdohzinyl, isoxazolyl, naphthyndinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1- oxidopyπdinyl, 1-oxidopynmidmyl, 1-oxidopyrazinyl, 1-oxidopyπdazmyl, 1 -phenyl- l//-pyrrolyl, phenazinyl, phenothiazmyl, phenoxazmyl, phthalazmyl, ptendmyl, purinyl, pyrrolyl, pyrazolyl, pyπdinyl, pyrazmyl, pyπmidmyl, pyπdazinyl, qmnazolmyl, qumoxalmyl, quinolmyl, quinuchdinyl, isoqumolmyl, tetrahydroqumolmyl, thiazolyl, ttuadiazolyl, tπazolyl, tetrazolyl, tπazmyl, and thiophenyl (i e , thienyl) Unless stated otherwise specifically in the specification, a heteroaryl group may be optionally substituted
'W-heteroaryl" refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical Unless stated otherwise specifically in the specification, an JV-heteroaryl group may be optionally substituted
"Heteroarylalkyl" refers to a radical of the formula -RbRf where Rb is an alkylene chain as defined above and Rf is a heteroaryl radical as defined above Unless stated otherwise specifically in the specification, a heteroarylalkyl group may be optionally substituted
The term "substituted" used herein means any of the above groups (z e , alkyl, alkylene, alkoxy, alkylammo, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, iV-heterocyclyl, heterocyclylalkyl, heteroaryl, Λ'-heteroaryl and/or heteroarylalkyl) wherein at least one hydrogen atom is replaced by a bond to a non-hydrogen atoms such as, but not limited to a halogen atom such as F, Cl, Br, and I, an oxygen atom in groups such as hydroxyl groups, alkoxy groups, and ester groups, a sulfur atom in groups such as thiol groups, thioalkyl groups, sulfone groups, sulfonyl groups, and sulfoxide groups, a nitrogen atom in groups such as amines, amides, alkylammes, dialkylamines, arylamines, alkylarylamines, diarylamines, N-oxides, lmides, and enamines, a silicon atom in groups such as tπalkylsilyl groups, dialkylarylsilyl groups, alkyldiarylsilyl groups, and tπarylsilyl groups, and other heteroatoms in vaπous other groups "Substituted" also means any of the above groups m which one or more hydrogen atoms are replaced by a higher-order bond (e g , a double- or triple-bond) to a heteroatom such as oxygen in oxo, carbonyl, carboxyl, and
9(5 ester groups, and nitrogen in groups such as lrmnes, oximes, hydrazones, and mtπles For example, "substituted" includes any of the above groups in which one or more hydrogen atoms are replaced with -NRgRh, -NRgC(=O)Rh, -NRgC(=O)NRgRh, -NRgC(-O)ORh, -NRgSO2Rh, -OC(-O)NRgRh, -ORg, -SRg, -SORE, -SO2R6, -OSO2R6, -SO2OR8, =NSO2Rg, and -SO2NRgRh "Substituted" also means any of the above groups in which one or more hydrogen atoms are replaced with -C(=0)Rg, -C(=O)ORg, -C(-O)NRgRh, -CH2SO2Rg, -CH2SO2NRgRh, -(CH2CH2O)2 ,0Rg In the foregoing, Rg and Rh are the same or different and independently hydrogen, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, N- heterocyclyl, heterocyclylalkyl, heteroaryl, iV-heteroaryl and/or heteroarylalkyl "Substituted" further means any of the above groups m which one or more hydrogen atoms are replaced by a bond to an amino, cyano, hydroxyl, lmino, mtro, oxo, thioxo, halo, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, Λf-heterocyclyl, heterocyclylalkyl, heteroaryl, iV-heteroaryl and/or heteroarylalkyl group In addition, each of the foregoing substituents may also be optionally substituted with one or more of the above substituents
"Prodrug" is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound of the invention Thus, the term "prodrug" refers to a metabolic precursor of a compound of the invention that is pharmaceutically acceptable A prodrug may be inactive when administered to a subject in need thereof, but is converted in vivo to an active compound of the invention Prodrugs are typically rapidly transformed in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, Bundgard, H , Design of Prodrugs (1985), pp 7-9, 21-24 (Elsevier, Amsterdam)) A discussion of prodrugs is provided in Higuchi, T , et al , A C S Symposium Seπes, VoI 14, and in Bioreversible Carriers in Drug Design, Ed Edward B Roche, American Pharmaceutical Association and Pergamon Press, 1987 The term "prodrug" is also meant to include any covalently bonded earners, which release the active compound of the invention in vivo when such prodrug is administered to a mammalian subject Prodrugs of a compound of the invention may be prepared by modifying functional groups present in the compound of the invention in such a way that the modifications are cleaved, either m routine manipulation or in vivo, to the parent compound of the invention Prodrugs include compounds of the invention wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the compound of the invention is administered to a mammalian subject, cleaves to form a free hydroxy, free ammo or free mercapto group, respectively Examples of prodrugs include, but are not limited to, acetate, formate and benzoate deπvatives of alcohol or amide derivatives of amine functional groups in the compounds of the invention and the like
The invention disclosed herein is also meant to encompass the in vivo metabolic products of the disclosed compounds Such products may result from, for example, the oxidation, reduction, hydrolysis, amidation, estenfication, and the like of the administered compound, primarily due to enzymatic processes Accordingly, the invention includes compounds produced by a process composing administering a compound of this invention to a mammal for a peπod of time sufficient to yield a metabolic product thereof Such products are typically identified by administering a radiolabeled compound of the invention in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to human, allowing sufficient time for metabolism to occur, and isolating its conversion products from the uπne, blood or other biological samples
"Stable compound" and "stable structure" are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of puπty from a reaction mixture, and formulation into an efficacious therapeutic agent "Optional" or "optionally" means that the subsequently descπbed event or circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not For example, "optionally substituted aryl" means that the aryl radical may or may not be substituted and that the descπption includes both substituted aryl radicals and aryl radicals having no substitution "Pharmaceutically acceptable earner, diluent or excipient" includes without limitation any adjuvant, earner, excipient, ghdant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals
"Pharmaceutically acceptable salt" includes both acid and base addition salts
"Pharmaceutically acceptable acid addition salt" refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but are not limited to, hydrochlonc acid, hydrobromic acid, sulfuric acid, nitric acid, phosphonc acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfomc acid, benzoic acid, 4-acetamidobenzoic acid, camphonc acid, camphor-10-sulfonic acid, capπc acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citnc acid, cyclamic acid, dodecylsulfunc acid, ethane- 1 ,2-disulfomc acid, ethanesulfomc acid, 2-hydroxyethanesulfonic acid, formic acid, fumaric acid, galactanc acid, gentisic acid, glucoheptonic acid, gluconic acid, glucuronic acid, glutamic acid, glutanc acid, 2-oxo-glutanc acid, glycerophosphonc acid, glycolic acid, hippunc acid, isobutync acid, lactic acid, lactobiomc acid, launc acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfomc acid, mucic acid, naphthalene-l,5-disulfomc acid, naphthalene-2-sulfonic acid, l-hydroxy-2-naphthoic acid, nicotinic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, propionic acid, pyroglutamic acid, pyruvic acid, salicylic acid, 4-aminosahcyhc acid, sebacic acid, steanc acid, succinic acid, tartanc acid, thiocyamc acid, />-toluenesulfomc acid, tnfluoroacetic acid, undecylemc acid, and the like
"Pharmaceutically acceptable base addition salt" refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable These salts are prepared from addition of an inorganic base or an organic base to the free acid Salts denved from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zmc, copper, manganese, aluminum salts and the like Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts Salts deπved from organic bases include, but are not limited to, salts of primary, S secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, lsopropylamme, tπmethylamme, diethylaimne, tπethylamme, tπpropylamme, diethanolamine, ethanolamme, deanol, 2-dimethylammoethanol,
2-diethylaminoethanol, dicyclohexylamine, lysine, argimne, histidine, caffeine,
10 procaine, hydrabamme, choline, betaine, benethamine, benzathine, ethylenediamme, glucosamine, methylglucamine, theobromine, tπethanolamine, tromethamine, purines, piperazme, pipeπdme, 7V-ethylpipeπdme, polyamme resms and the like Particularly preferred organic bases are lsopropylamme, diethylamme, ethanolamme, tπmethylamine, dicyclohexylamine, choline and caffeine
I5 Often crystallizations produce a solvate of the compound of the invention As used herein, the term "solvate" refers to an aggregate that compπses one or more molecules of a compound of the invention with one or more molecules of solvent The solvent may be water, in which case the solvate may be a hydrate Alternatively, the solvent may be an organic solvent Thus, the compounds of the
20 present invention may exist as a hydrate, including a monohydrate, dihydrate, hermhydrate, sesquihydrate, tnhydrate, tetrahydrate and the like, as well as the corresponding solvated forms The compound of the invention may be true solvates, while in other cases, the compound of the invention may merely retain adventitious water or be a mixture of water plus some adventitious solvent
25 A "pharmaceutical composition" refers to a formulation of a compound of the invention and a medium generally accepted m the art for the delivery of the biologically active compound to mammals, e g , humans Such a medium includes all pharmaceutically acceptable earners, diluents or excipients therefor
The compounds of the invention, or their pharmaceutically acceptable
30 salts may contain one or more asymmetric centers and may thus give πse to enantiomers, diastereomers, and other stereoisomeπc forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for ammo acids The present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms Optically active (+) and (-), (R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or deπvative) using, for example, chiral high pressure liquid chromatography (HPLC) When the compounds descπbed herein contain olefinic double bonds or other centres of geometnc asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometnc isomers Likewise, all tautomeπc forms are also intended to be included
A "stereoisomer" refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable The present invention contemplates vaπous stereoisomers and mixtures thereof and includes "enantiomers", which refers to two stereoisomers whose molecules are nonsupeπmposeable mirror images of one another
A "tautomer" refers to a proton shift from one atom of a molecule to another atom of the same molecule The present invention includes tautomers of any said compounds
In accordance with the present disclosure, the compounds descnbed herein are designed to be substantially active or localized in the gastrointestinal lumen of a human or animal subject The term "gastrointestinal lumen" is used interchangeably herein with the term "lumen," to refer to the space or cavity within a gastrointestinal tract (GI tract, which can also be referred to as the gut), delimited by the apical membrane of GI epithelial cells of the subject In some embodiments, the compounds are not absorbed through the layer of epithelial cells of the GI tract (also known as the GI epithelium) "Gastrointestinal mucosa" refers to the layer(s) of cells separating the gastrointestinal lumen from the rest of the body and includes gastric and intestinal mucosa, such as the mucosa of the small intestine A "gastrointestinal epithelial cell" or a "gut epithelial cell" as used herein refers to any epithelial cell on the surface of the gastrointestinal mucosa that faces the lumen of the gastrointestinal tract, including, for example, an epithelial cell of the stomach, an intestinal epithelial cell, a colonic epithelial cell, and the like
"Substantially systemically non-bioavailable" and/or "substantially 5 impermeable" as used herein (as well as vaπations thereof) generally refer to situations in which a statistically significant amount, and in some embodiments essentially all of the compound of the present disclosure (which includes the NHE-inhibitor small molecule), remains in the gastrointestinal lumen For example, m accordance with one or more embodiments of the present disclosure, preferably at least about 70%, about
10 80%, about 90%, about 95%, about 98%, about 99%, or even about 99 5%, of the compound remains in the gastrointestinal lumen In such cases, localization to the gastrointestinal lumen refers to reducing net movement across a gastrointestinal layer of epithelial cells, for example, by way of both transcellular and paracellular transport, as well as by active and/or passive transport The compound m such embodiments is
I5 hindered from net permeation of a layer of gastrointestinal epithelial cells in transcellular transport, for example, through an apical membrane of an epithelial cell of the small intestine The compound in these embodiments is also hindered from net permeation through the "tight junctions" in paracellular transport between gastrointestinal epithelial cells lining the lumen
20 In this regard it is to be noted that, in one particular embodiment, the compound is essentially not absorbed at all by the GI tract or gastrointestinal lumen As used herein, the terms "substantially impermeable" or "substantially systemically non- bioavailable" refers to embodiments wherein no detectable amount of absorption or permeation or systemic exposure of the compound is detected, using means generally
25 known in the art
In this regard it is to be further noted, however, that in alternative embodiments "substantially impermeable" or "substantially systemically non bioavailable" provides or allows for some limited absorption in the GI tract, and more particularly the gut epithelium, to occur (e g , some detectable amount of absorption,
30 such as for example at least about 0 1%, 0 5%, 1% or more and less than about 30%, 20%, 10%, 5%, etc , the range of absorption being for example between about 1% and 30%, or 5% and 20%, etc , stated another way, "substantially impermeable" or "substantially systemically non bioavailable" refers to compounds that exhibit some detectable permeability to an epithelium layer of cells in the GI tract of less than about 20% of the administered compound (e g , less than about 15%, about 10%, or even about 5%, and for example greater than about 0 5%, or 1%), but then are cleared by the liver (i e , hepatic extraction) and/or the kidney (i e , renal excretion)
B. Permeability
In this regard it is to be noted that, in vaπous embodiments, the ability of the compound to be substantially systemically non-bioavailable is based on the compound charge, size, and/or other physicochemical parameters (e g , polar surface area, number of hydrogen bond donors and/or acceptors therein, number of freely rotatable bonds, etc ) More specifically, it is to be noted that the absorption character of a compound can be selected by applying principles of pharmacodynamics, for example, by applying Lipinski's rule, also known as "the rule of five " Although not a rule, but rather a set of guidelines, Lipmski shows that small molecule drugs with (i) a molecular weight, (ii) a number of hydrogen bond donors, (in) a number of hydrogen bond acceptors, and/or (iv) a water/octanol partition coefficient (Moπguchi Log P), greater than a certain threshold value, generally do not show significant systemic concentration (i e , are generally not absorbed to any significant degree) (See, e g , Lipmski et al , Advanced Drug Delivery Reviews, 46, 2001 3-26, incorporated herein by reference ) Accordingly, substantially systemically non-bioavailable compounds (e g , substantially systemically non-bioavailable NHE inhibitor compounds) can be designed to have molecular structures exceeding one or more of Lipinski's threshold values (See also Lipmski et al , Experimental and Computational Approaches to Estimate Solubility and Permeability in Drug Discovery and Development Settings, Adv Drug Delivery Reviews, 46 3-26 (2001), and Lipmski, Drug-like Properties and the Causes of Poor Solubility and Poor Permeability, J Pharm & Toxicol Methods, 44235-249 (2000), incorporated herein by reference ) In some embodiments, for example, a substantially impermeable or substantially systemically non-bioavailable NHE inhibitor compound of the present disclosure can be constructed to feature one or more of the following characteπstics (i) a MW greater than about 500 Da, about 1000 Da, about 2500 Da, about 5000 Da, about 10,000 Da or more (in the non-salt form of the compound), (ii) a total number of NH and/or OH and/or other potential hydrogen bond donors greater than about 5, about 10, about 15 or more, (m) a total number of O atoms and/or N 5 atoms and/or other potential hydrogen bond acceptors greater than about 5, about 10, about 15 or more, and/or (iv) a Monguchi partition coefficient greater than about 105 (i e , Log P greater than about 5, about 6, about 7, etc ), or alternatively less than about 10 (i e , a Log P of less than 1, or even 0)
In view of the foregoing, and as previously noted herein, essentially any
10 known NHE inhibitor small molecule (descnbed herein and/or in the art) can be used in designing a substantially systemically non bioavailable NHE inhibitor molecular structure, in accordance with the present disclosure In addition to the parameters noted above, the molecular polar surface area (i e , "PSA"), which may be characterized as the surface belonging to polar atoms, is a descπptor that has also been shown to correlate
15 well with passive transport through membranes and, therefore, allows prediction of transport properties of drugs It has been successfully applied for the prediction of intestinal absorption and Caco2 cell monolayer penetration (For Caco2 cell monolayer penetration test details, see for example the descπption of the Caco2 Model provided in Example 31 of U S Pat No 6,737,423, the entire contents of which are incorporated
20 herein by reference for all relevant and consistent purposes, and the text of Example 31 in particular, which may be applied for example to the evaluation or testing of the compounds of the present disclosure ) PSA is expressed in A2 (squared angstroms) and is computed from a three dimensional molecular representation A fast calculation method is now available (see, e g , Ertl et al , Journal of Medicinal Chemistry, 2000, 43,
25 3714-3717, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes) using a desktop computer and commercially available chemical graphic tools packages, such as ChemDraw The term "topological PSA" (tPSA) has been coined for this fast-calculation method tPSA is well correlated with human absorption data with common drugs (see, e g , Table 2, below)
30
Table 2 name % FA" TPSA6 metoprolol 102 50 7 nordiazepam 99 41 5 diazepam 97 32 7 oxprenolol 97 50 7 phenazone 97 26.Θ oxazepam 97 Gl 7 alprenolol 96 41 9 practolel 95 70.6 pindolol 92 57.3 clpiofloxtidn 69 74 0 meiolazone 64 92 5 traπexamic acid 55 03 3 atenolol 54 84 0 sulpiride 101 7 rmmittol 2G 121 4 foscarnet 17 94 8 sulfasalazine 12 141 3 olsalazine 2 3 139 8 lactulose 06 IU7 4 raffinose 03 208 7
(from Ertl et al., J. Med. Chem., 2000, 43:3714-3717). Accordingly, in some preferred embodiments, the compounds of the present disclosure may be constructed to exhibit a tPSA value greater than about 100 A2, about 120 A2, about 130 A2, or about 140 A2, and in some instances about 150 A2, about 200 A2, about 250 A2, about 270 A2, about 300 A2, about 400 A2,or even about 500 A2, such that the compounds are substantially impermeable or substantially systemically non-bioavailable (as defined elsewhere herein).
Because there are exceptions to Lipinski's "rule," or the tPSA model, the permeability properties of the compounds of the present disclosure may be screened experimentally. The permeability coefficient can be determined by methods known to those of skill in the art, including for example by Caco-2 cell permeability assay and/or using an artificial membrane as a model of a gastrointestinal epithelial cell. (As previously noted above, see for example U.S. Patent No. 6,737,423, Example 31 for a description of the Caco-2 Model, which is incorporated herein by reference). A synthetic membrane impregnated with, for example, lecithin and/or dodecane to mimic the net permeability characteristics of a gastrointestinal mucosa, may be utilized as a model of a gastrointestinal mucosa. The membrane can be used to separate a compartment containing the compound of the present disclosure from a compartment where the rate of permeation will be monitored. Also, parallel artificial membrane permeability assays (PAMPA) can be performed Such in vitro measurements can reasonably indicate actual permeability in vivo (See, for example, Wohnsland et al , J Med Chem , 2001, 44 923-930, Schmidt et al , Milhpore Corp Application Note, 2002, n° ANl 725EN00, and n0 ANl 728EN00, incorporated herein by reference ) Accordingly, m some embodiments, the compounds utilized in the methods of the present disclosure may have a permeability coefficient, Papp, of less than about 100 x 106 cm/s, or less than about 10 x 106 cm/s, or less than about 1 x 106 cm/s, or less than about 0 1 x 106 cm/s, when measured using means known in the art (such as for example the permeability experiment descπbed m Wohnsland et al , J Med Chem , 2001, 44 923-930, the contents of which is incorporated herein by reference)
As previously noted, in accordance with the present disclosure, NHE inhibitor small molecules are modified as descπbed above to hinder the net absorption through a layer of gut epithelial cells, rendeπng them substantially systemically non- bioavailable In some particular embodiments, the compounds of the present disclosure compπse an NHE-mhibiting small molecule linked, coupled or otherwise attached to a moiety Z, which may be an oligomer moiety, a polymer moiety, a hydrophobic moiety, a hydrophilic moiety, and/or a charged moiety, which renders the overall compound substantially impermeable or substantially systemically non bioavailable In some preferred embodiments, the NHE-inhibiting small molecule is coupled to a multimer or polymer portion or moiety, such that the resulting NHE-Z molecule is substantially impermeable or substantially systemically non bioavailable The multimer or polymer portion or moiety may be of a molecular weight greater than about 500 Daltons (Da), about 1000 Da, about 2500 Da, about 5000 Da, about 10,000 Da or more, and in particular may have a molecular weight in the range of about 1000 Daltons (Da) to about 500,000 Da, preferably in the range of about 5000 to about 200,000 Da, and more preferably may have a molecular weight that is sufficiently high to essentially preclude any net absorption through a layer of gut epithelial cells of the compound For example, an NHE-mhibitmg small molecule may be linked to at least one repeat unit of a polymer portion or moiety according, for example, to the structure of Formula (XIIA) or Formula (XIIB), as illustrated herein In these or other particular embodiments, the NHE-inhibiting small molecule is modified as descπbed herein to substantially hinder its net absorption through a layer of gut epithelial cells and may compπse, for example, a NHE-inhibitmg compound linked, coupled or otherwise attached to a substantially impermeable or substantially systemically non-bioavailable "Core" moiety, as descπbed above
C. Persistent Inhibitory Effect
In other embodiments, the substantially impermeable or substantially systemically non-bioavailable NHE-mhibitmg compounds utilized in the treatment methods of the present disclosure may additionally exhibit a persistent inhibitor effect This effect manifests itself when the inhibitory action of a compound at a certain concentration in equilibrium with the epithelial cell (e g , at or above its inhibitory concentration, IC) does not revert to baseline (i e , sodium transport without inhibitor) after the compound is depleted by simple washing of the luminal content This effect can be interpreted as a result of the tight binding of the NHE- mhibitmg compounds to the NHE protein at the intestinal apical side of the gut epithelial cell The binding can be considered as quasi-irreversible to the extent that, after the compound has been contacted with the gut epithelial cell and subsequently washed off said gut epithelial cell, the flux of sodium transport is still significantly lower than m the control without the compound This persistent inhibitory effect has the clear advantage of maintaining drug activity within the GI tract even though the residence time of the active m the upper GI tract is short, and when no entero-bihary recycling process is effective to replenish the compound concentration near its site of action Such a persistent inhibitory effect has an obvious advantage in terms of patient compliance, but also in limiting drug exposure within the GI tract
The persistence effect can be determined using in vitro methods, in one instance, cell lines expressing NHE transporters are split in different vials and treated with a NHE inhibiting compound and sodium solution to measure the rate of sodium uptake The cells m one set of vials are washed for different penods of time to remove the inhibitor, and sodium uptake measurement is repeated after the washing Compounds that maintain their inhibitory effect after multiple/lengthy washing steps (compared to the inhibitory effect measured in the vials where washing does not occur) are persistent inhibitors Persistence effect can also be characterized ex vivo by using the everted sac technique, whereby transport of Na is monitored using an excised segment of GI perfused with a solution containing the inhibitor and shortly after flushing the bathing solution with a buffer solution free from inhibitor A persistence effect can also be characteπzed in vivo by observing the time needed for sodium balance to return to normal when the inhibitor treatment is discontinued The limit of the method resides in the fact that apical cells (and therefore apical NHE transporters) are sloughed off after a period of 3 to 4 days, the typical turnover time of gut epithelial cells A persistence effect can be achieved by increasing the residence time of the active compound at the apical surface of the gut epithelial cells, this can be obtained by designing NHE antiport inhibitors with several NHE inhibiting moieties built-m the small molecule or oligomer (wherein "several" as used herein typically means at least about 2, about 4, about 6 or more) Examples of such structures in the context of analogs of the antibiotic vancomycin are given in Griffin, et al , J Am Chem Soc , 2003, 125, 6517-6531 Alternatively the compound compπses groups that contnbute to increase the affinity towards the gut epithelial cell so as to increase the time of contact with the gut epithelial cell surface Such groups are referred to as being "mucoadhesive " More specifically, the Core or L moiety can be substituted by such mucoadhesive groups, such as polyacrylates, partially deacetylated chitosan or polyalkylene glycol (See also Patil, S B et al , Curr Drug Dehv , 2008, Oct 5(4), pp 312-8 )
D. GI Enzyme Resistance
Because the compounds utilized in the treatment methods of the present disclosure are preferably substantially systemically non-bioavailable, and/or preferably exhibit a persistent inhibitory effect, it is also desirable that, duπng their prolonged residence time in the gut, these compounds sustain the hydrolytic conditions prevailing in the upper GI tract In such embodiments, compounds of the present disclosure are resistant to enzymatic metabolism For example, administered compounds are preferably resistant to the activity of P450 enzymes, glucurosyl transferases, sulfotransferases, glutathione S-transferases, and the like, in the intestinal mucosa, as well as gastπc (e g , gastric lipase, and pepsme), pancreatic (e g , trypsin, triglyceride pancreatic lipase, phosphohpase A2, endonucleases, nucleotidases, and alpha-amylase), and brush-border enzymes (e g , alkaline phosphatase, glycosidases, and proteases) generally known in the art
The compounds that are utilized in methods of the present disclosure are also preferably resistant to metabolism by the bacteπal flora of the gut, that is, the compounds are not substrates for enzymes produced by bacteπal flora In addition, the compounds administered in accordance with the methods of the present disclosure may be substantially inactive towards the gastrointestinal flora, and do not disrupt bacteπal growth or survival As a result, m vaπous embodiments herein, the minimal inhibitory concentration (or "MIC") against GI flora is desirably greater than about 15 μg/ml, about 30 μg/ml, about 60 μg/ml, about 120 μg/ml, or even about 240 μg/ml, the MIC in vanous embodiments being for example between about 16 and about 32 μg/ml, or between about 64 and about 128 μg/ml, or greater than about 256 μg/ml
To one skilled in the art of medicinal chemistry, metabolic stability can be achieved in a number of ways Functionality susceptible to P450-mediated oxidation can be protected by, for example, blocking the point of metabolism with a halogen or other functional group Alternatively, electron withdrawing groups can be added to a conjugated system to generally provide protection to oxidation by reducing the electrophilicity of the compound Proteolytic stability can be achieved by avoiding secondary amide bonds, or by incorporating changes in stereochemistry or other modifications that prevent the drug from otherwise being recognized as a substrate by the metabolizing enzyme
E. Sodium and/or Fluid Output
It is also to be noted that, in various embodiments of the present disclosure, one or more of the NHE-Z inhibiting compounds (monovalent or divalent) detailed herein, when administered either alone or in combination with one or more additional pharmaceutically active compounds or agents (including, for example, a fluid-absorbing polymer) to a patient in need thereof, may act to increase the patient's daily fecal output of sodium by at least about 20, about 30 mmol, about 40 mmol, about 50 mmol, about 60 mmol, about 70 mmol, about 80 mmol, about 90 mmol, about 100 mmol, about 125 mmol, about 150 mmol or more, the increase being for example within the range of from about 20 to about 150 mmol/day, or from about 25 to about 100 mmol/day, or from about 30 to about 60 mmol/day
Additionally, or alternatively, it is also to be noted that, in various embodiments of the present disclosure, one or more of the NHE-Z inhibiting compounds (monovalent or divalent) detailed herein, when administered either alone or in combination with one or more additional pharmaceutically active compounds or agents (including, for example, a fluid-absorbing polymer) to a patent in need thereof, may act to increase the patient's daily fluid output by at least about 100 ml, about 200 ml, about 300 ml, about 400 ml, about 500 ml, about 600 ml, about 700 ml, about 800 ml, about 900 ml, about 1000 ml or more, the increase being for example within the range of from about 100 to about 1000 ml/day, or from about 150 to about 750 ml/day, or from about 200 to about 500 ml/day (assuming isotonic fluid)
F. Craax and IC50
It is also to be noted that, in various embodiments of the present disclosure, one or more of the NHE-Z inhibiting compounds (monovalent or divalent) detailed herein, when administered either alone or in combination with one or more additional pharmaceutically active compounds or agents (including, for example, a fluid-absorbing polymer) to a patient in need thereof at a dose resulting in at least a
10% increase in fecal water content, has a Craax that is less than the IC50 for NHE-3, more specifically, less than about 1OX (10 times) the IC50, and, more specifically still, less than about IOOX (100 times) the IC50
Additionally, or alternatively, it is also to be noted that, in various embodiments of the present disclosure, one or more of the NHE-Z inhibiting compounds (monovalent or divalent) detailed herein, when administered either alone or in combination with one or more additional pharmaceutically active compounds or agents (including, for example, a fluid-absorbing polymer) to a patient in need thereof, may have a Cmax of less than about 10 ng/ml, about 7 5 ng/ml, about 5 ng/ml, about 2 5 ng/ml, about 1 ng/ml, or about 0 5 ng/ml, the Cm8x bemg for example within the range of about 1 ng/ml to about 10 ng/ml, or about 2 5 ng/ml to about 7 5 ng/ml
Additionally, or alternatively, it is also to be noted that, in various embodiments of the present disclosure, one or more of the NHE-Z inhibiting compounds (monovalent or divalent) detailed herein, when administered either alone or in combination with one or more additional pharmaceutically active compounds or agents (including, for example, a fluid-absorbing polymer) to a patient in need thereof, may have a IC5O of less than about 10 μM, about 7 5 μM, about 5 μM, about 2 5 μM, about 1 μM, or about 0 5 μM, the IC50 being for example withm the range of about 1 μM to about 10 μM, or about 2 5 μM to about 7 5 μM
Additionally, or alternatively, it is also to be noted that, in various embodiments of the present disclosure, one or more of the NHE-Z inhibiting compounds (monovalent or divalent) detailed herein, when administered to a patient in need thereof, may have a ratio of IC50 Cmax, wherein IC50 and Cmax are expressed in terms of the same units, of at least about 10, about 50, about 100, about 250, about 500, about 750, or about 1000
Additionally, or alternatively, it is also to be noted that, in vaπous embodiments of the present disclosure, wherein one or more of the NHE-Z inhibiting compounds (monovalent or divalent) as detailed herein is orally administered to a patent in need thereof, within the therapeutic range or concentration, the maximum compound concentration detected m the serum, defined as Cmax, is lower than the NHE inhibitory concentration IC50 of said compound As previously noted, as used herein, IC50 is defined as the quantitative measure indicating the concentration of the compound required to inhibit 50% of the NHE-mediated Na / H antiport activity in a cell based assay
IV. Pharmaceutical Compositions and Methods of Treatment
A. Compositions and Methods 1. Fluid Retention and/or Salt Overload Disorders A pharmaceutical composition or preparation that may be used in accordance with the present disclosure for the treatment of various disorders associated with fluid retention and/or salt overload in the gastrointestinal tract (e g , hypertension, heart failure (in particular, congestive heart failure), chrome kidney disease, end-stage renal disease, liver disease and/or peroxisome prohferator-activated receptor (PPAR) gamma agonist-induced fluid retention) compπses, in general, the substantially impermeable or substantially systemically non-bioavailable NHE-inhibiting compound of the present disclosure, as well as various other optional components as further detailed herein below (e g , pharmaceutically acceptable excipients, etc ) The compounds utilized in the treatment methods of the present disclosure, as well as the pharmaceutical compositions comprising them, may accordingly be administered alone, or as part of a treatment protocol or regiment that includes the administration or use of other beneficial compounds (as further detailed elsewhere herein) In some particular embodiments, the NHE-inhibitmg compound, including any pharmaceutical composition compπsing the compound, is administered with a fluid-absorbmg polymer (as more fully descπbed below)
A "subject" or "mammal" is preferably a human, but can also be an animal in need of treatment with a compound of the disclosure, e g , companion animals (e g , dogs, cats, and the like), farm animals (e g , cows, pigs, horses and the like) and laboratory animals (e g , rats, mice, guinea pigs and the like)
Subjects "in need of treatment" with a compound of the present disclosure, or subjects "in need of NHE inhibition" include subjects with diseases and/or conditions that can be treated with substantially impermeable or substantially systemically non-bioavailable NHE-inhibitmg compounds, with or without a fluid- absorbing polymer, to achieve a beneficial therapeutic and/or prophylactic result A beneficial outcome includes a decrease in the seventy of symptoms or delay in the onset of symptoms, increased longevity and/or more rapid or more complete resolution of the disease or condition For example, a subject in need of treatment may be suffering from hypertension, from salt-sensitive hypertension which may result from dietary salt intake, from a πsk of a cardiovascular disorder (e g , myocardial infarction, congestive heart failure and the like) resulting from hypertension, from heart failure (e g , congestive heart failure) resulting m fluid or salt overload, from chronic kidney disease resulting m fluid or salt overload, from end stage renal disease resulting in fluid or salt overload, from liver disease resulting in fluid or salt overload, from peroxisome prohferator-activated receptor (PPAR) gamma agonist-induced fluid retention, or from edema resulting from congestive heart failure or end stage renal disease In various embodiments, a subject in need of treatment typically shows signs of hypervolemia resulting from salt and fluid retention that are common features of congestive heart failure, renal failure or liver cirrhosis Fluid retention and salt retention manifest themselves by the occurrence of shortness of breath, edema, ascites or mterdialyhc weight gain Other examples of subjects that would benefit from the treatment are those suffering from congestive heart failure and hypertensive patients and, particularly, those who are resistant to treatment with diuretics, i e , patients for whom very few therapeutic options are available A subject "m need of treatment" also includes a subject with hypertension, salt-sensitive blood pressure and subjects with systolic / diastolic blood pressure greater than about 130-139 / 85-89 mm Hg
Administration of NHE inhibitors, with or without administration of fluid-absorbing polymers, may be beneficial for patients put on "non added salt" dietary regimen (i e , 60-100 mmol of Na per day), to liberalize their diet while keeping a neutral or slightly negative sodium balance (i e , the overall uptake of salt would be equal of less than the secreted salt) In that context, "liberalize their diet" means that patients treated may add salt to their meals to make the meals more palatable, or/and diversify their diet with salt-containing foods, thus maintaining a good nutntional status while improving their quality of life
The treatment methods descπbed herein may also help patients with edema associated with chemotherapy, pre menstrual fluid overload and preeclampsia (pregnancy-induced hypertension)
Accordingly, it is to be noted that the present disclosure is further directed to methods of treatment involving the administration of the compound of the present disclosure, or a pharmaceutical composition comprising such a compound Such methods may include, for example, a method for treating hypertension, the method comprising administering to the patient a substantially impermeable or substantially systemically non-bioavailable NHE-inhibiting compound, or a composition compnsing it The method may be for reducing fluid overload associated with heart failure (in particular, congestive heart failure), the method comprising administering to the patient a substantially impermeable or substantially systemically 5 non-bioavailable NHE-inhibiting compound or pharmaceutical composition compnsing it The method may be for reducing fluid overload associated with end stage renal disease, the method compnsing administenng to the patient a substantially impermeable or substantially systemically non-bioavailable NHE-inhibitmg compound or composition compnsing it The method may be for reducing fluid overload associated
10 with peroxisome proliferator-activated receptor (PPAR) gamma agonist therapy, the method compnsing administenng to the patient a substantially impermeable or substantially systemically non-bioavailable NHE-inhibiting compound or composition compnsing it Additionally, or alternatively, the method may be for decreasing the activity of an intestinal NHE transporter in a patient, the method compnsmg
I5 administenng to the patient a substantially impermeable or substantially systemically non-bioavailable NHE-inhibiting compound, or a composition compnsing it
2. Gastrointestinal Tract Disorders
A pharmaceutical composition or preparation that may be used m
20 accordance with the present disclosure for the treatment of vanous gastrointestinal tract disorders, including the treatment or reduction of pain associated with gastrointestinal tract disorders, comprises, in general, any small molecule, which may be monovalent or polyvalent, that is effective or active as an NHE-inhibitor and that is substantially active in the GI tract, in particular, the substantially impermeable or substantially systemically
25 non-bioavailable NHE-inhibiting compound of the present disclosure, as well as vanous other optional components as further detailed herein below (e g , pharmaceutically acceptable excipients, etc ) The compounds utilized in the treatment methods of the present disclosure, as well as the pharmaceutical compositions compnsing them, may accordingly be administered alone, or as part of a treatment protocol or regiment that
30 includes the administration or use of other beneficial compounds (as further detailed elsewhere herein) In some particular embodiments, the NHE-inhibitmg compound, including any pharmaceutical composition composing the compound, is administered with a fluid-absorbing polymer (as more fully descπbed below)
A "subject" is preferably a human, but can also be an animal m need of treatment with a compound of the disclosure, e g , companion animals (e g , dogs, cats, and the like), farm animals (e g , cows, pigs, horses and the like) and laboratory animals (e g , rats, mice, guinea pigs and the like)
Subjects "m need of treatment" with a compound of the present disclosure, or subjects "in need of NHE inhibition" include subjects with diseases and/or conditions that can be treated with substantially impermeable or substantially systemically non-bioavailable NHE-mhibiting compounds, with or without a fluid- absorbmg polymer, to achieve a beneficial therapeutic and/or prophylactic result A beneficial outcome includes a decrease in the seventy of symptoms or delay m the onset of symptoms, increased longevity and/or more rapid or more complete resolution of the disease or condition For example, a subject in need of treatment is suffenng from a gastrointestinal tract disorder, the patient is suffenng from a disorder selected from the group consisting of a gastrointestinal motility disorder, irntable bowel syndrome, chronic constipation, chrome idiopathic constipation, chronic constipation occurnng in cystic fibrosis patients, chronic constipation occurnng in chronic kidney disease patients, calcium-induced constipation in osteoporotic patients, opioid-mduced constipation, a functional gastrointestinal tract disorder, gastroesophageal reflux disease, functional heartburn, dyspepsia, functional dyspepsia, non-ulcer dyspepsia, gastroparesis, chronic intestinal pseudo-obstruction, Crohn's disease, ulcerative colitis and related diseases referred to as inflammatory bowel syndrome, colonic pseudoobstruction, and the like In vanous preferred embodiments, the constipation to be treated is associated with the use of a therapeutic agent, associated with a neuropathic disorder, post-surgical constipation (postoperative ileus), associated with a gastrointestinal tract disorder, idiopathic (functional constipation or slow transit constipation), associated with neuropathic, metabolic or endocnne disorder (e g , diabetes mellitus, renal failure, hypothyroidism, hyperthyroidism, hypocalcaemia, Multiple Sclerosis, Parkinson's disease, spinal cord lesions, neurofibromatosis, autonomic neuropathy, Chagas disease, Hirschsprung's disease or cystic fibrosis, and the like) Constipation may also be the result of surgery (postoperative ileus) or due the use of drugs such as analgesics (e g , opioids), antihypertensives, anticonvulsants, antidepressants, antispasmodics and antipsychotics Accordingly, it is to be noted that the present disclosure is further directed to methods of treatment involving the administration of the compound of the present disclosure, or a pharmaceutical composition compπsmg such a compound Such methods may include, for example, a method for increasing gastrointestinal motility in a patient, the method compπsmg administering to the patient a substantially non-permeable or substantially non-bioavailable NHE-inhibitmg compound, or a composition comprising it Additionally, or alternatively, the method may be for decreasing the activity of an intestinal NHE transporter in a patient, the method comprising administering to the patient a substantially non-permeable or substantially non-bioavailable NHE-mhibiting compound, or a composition comprising it Additionally, or alternatively, the method may be for treating a gastrointestinal tract disorder, a gastrointestinal motility disorder, irritable bowel syndrome, chronic calcium- mduced constipation in osteoporotic patients, chronic constipation occurring in cystic fibrosis patients, chronic constipation occurring in chronic kidney disease patients, a functional gastrointestinal tract disorder, gastroesophageal reflux disease, functional heartburn, dyspepsia, functional dyspepsia, non-ulcer dyspepsia, gastroparesis, chrome intestinal pseudo-obstruction, colonic pseudo-obstruction, Crohn's disease, ulcerative colitis, inflammatory bowel disease, the method compπsing administering an antagonist of the intestinal NHE, and more specifically a substantially non-permeable or substantially non-bioavailable NHE-mhibiting compound, or composition, either orally or by rectal suppository Additionally, or alternatively, the method may be for treating or reducing pain, including visceral pain, pain associated with a gastrointestinal tract disorder or pain associated with some other disorder, the method compπsing admmisteπng to a patient a substantially non-permeable or substantially non- bioavailable NHE-inhibiting compound, or composition Additionally, or alternatively, the method may be for treating inflammation, including inflammation of the gastrointestinal tract, e g , inflammation associated with a gastrointestinal tract disorder or infection or some other disorder, the method comprising administering to a patient a substantially non-permeable or substantially non-bioavailable NHE-inhibiting compound, or composition
B. Combination Therapies
1. Fluid Retention and/or Salt Overload Disorders
As previously noted, the compounds described herein can be used alone or in combination with other agents For example, the compounds can be administered together with a diuretic (i e , High Ceiling Loop Diuretics, Benzothiadiazide Diuretics, Potassium Sparing Diuretics, Osmotic Diuretics), cardiac glycoside, ACE inhibitor, angiotensin-2 receptor antagonist, calcium channel blocker, beta blocker, alpha blocker, central alpha agonist, vasodilator, blood thinner, anti-platelet agent, hpid-loweπng agent, peroxisome prohferator-activated receptor (PPAR) gamma agonist agent or compound or with a fluid-absorbing polymer as more fully descπbed below The agent can be covalently attached to a compound descπbed herein or it can be a separate agent that is administered together with or sequentially with a compound descnbed herein in a combination therapy
Combination therapy can be achieved by administering two or more agents, e g , a substantially non-permeable or substantially systemically non- bioavailable NHE-inhibiting compound descπbed herein and a diuretic, cardiac glycoside, ACE inhibitor, angiotensm-2 receptor antagonist, calcium channel blocker, beta blocker, alpha blocker, central alpha agonist, vasodilator, blood thinner, antiplatelet agent or compound, each of which is formulated and administered separately, or by admmistenng two or more agents in a single formulation Other combinations are also encompassed by combination therapy For example, two agents can be formulated together and administered in conjunction with a separate formulation containing a third agent While the two or more agents in the combination therapy can be administered simultaneously, they need not be For example, administration of a first agent (or combination of agents) can precede administration of a second agent (or combination of agents) by minutes, hours, days, or weeks Thus, the two or more agents can be administered withm minutes of each other or within 1, 2, 3, 6, 9, 12, 15, 18, or 24 hours of each other or withm 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14 days of each other or within 2, 3, 4, 5, 6, 7, 8, 9, or weeks of each other In some cases even longer intervals are possible While in many cases it is desirable that the two or more agents used in a combination therapy be present in within the patient's body at the same time, this need not be so
Combination therapy can also include two or more administrations of one or more of the agents used in the combination For example, if agent X and agent Y are used m a combination, one could administer them sequentially in any combination one or more times, e g , in the order X-Y-X, X-X-Y, Y-X-Y, Y Y-X, X-X-Y-Y, etc The compounds descπbed herein can be used in combination therapy with a diuretic Among the useful analgesic agents are, for example High Ceiling Loop Diuretics [Furosemide (Lasix), Ethacrynic Acid (Edecnn) ,Bumetamde (Bumex)], Benzothiadiazide Diuretics [Hydrochlorothiazide (Hydrodmπl), Chlorothiazide (Diuπl), Clorthahdone (Hygroton), Benzthiazide (Aguapres), Bendroflumethiazide (Naturetin), Methyclothiazide (Aguatensen), Polythiazide (Renese), Indapamide (Lozol), Cyclothiazide (Anhydron), Hydroflumethiazide (Diucardra), Metolazone (Dmlo), Qmnethazone (Hydromox), Tπchlormethiazide (Naqua)], Potassium Sparing Diuretics [Spironolactone (Aldactone), Tπamterene (Dyrenmm), Amiloπde (Midamor)], and Osmotic Diuretics [Manmtol (Osmitrol)] Diuretic agents in the various classes are known and descπbed in the literature
Cardiac glycosides (cardenohdes) or other digitalis preparations can be administered with the compounds of the disclosure in co therapy Among the useful cardiac glycosides are, for example Digitoxm (Crystodigin), Digoxin (Lanoxin) or Deslanoside (Cedilamd-D) Cardiac glycosides in the vaπous classes are descπbed in the literature
Angiotensin Converting Enzyme Inhibitors (ACE Inhibitors) can be administered with the compounds of the disclosure in co-therapy Among the useful ACE inhibitors are, for example Captopπl (Capoten), Enalapπl (Vasotec), Lisinopπl (Pπnivil) ACE inhibitors in the vaπous classes are descπbed in the literature Angiotensin-2 Receptor Antagonists (also referred to as ATi -antagonists or angiotensin receptor blockers, or ARB 's) can be administered with the compounds of the disclosure in co-therapy Among the useful Angiotensm-2 Receptor Antagonists are, for example Candesartan (Atacand), Eprosartan (Teveten), Irbesartan (Avapro), Losartan (Cozaar), Telmisartan (Micardis), Valsartan (Diovan) Angiotensm-2 Receptor Antagonists in the various classes are descπbed in the literature Calcium channel blockers such as Amlodipme (Norvasc, Lotrel),
Bepπdil (Vascor), Diltiazem (Cardizem, Tiazac), Felodipine (Plendil), Nifedipine (Adalat, Procardia), Nimodipine (Nimotop), Nisoldipme (Sular), Verapamil (Calan, Isoptin, Verelan) and related compounds descπbed in, for example, EP 625162B1, U S Pat No 5,364,842, U S Pat No 5,587,454, U S Pat No 5,824,645, U S Pat No 5,859,186, U S Pat No 5,994,305, U S Pat No 6,087,091, U S Pat No 6,136,786, WO 93/13128 Al, EP 1336409 Al, EP 835126 Al, EP 835126 Bl, U S Pat No 5,795,864, U S Pat No 5,891,849, U S Pat No 6,054,429, WO 97/01351 Al, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes, can be used with the compounds of the disclosure Beta blockers can be administered with the compounds of the disclosure in co- therapy Among the useful beta blockers are, for example Acebutolol (Sectral), Atenolol (Tenormin), Betaxolol (Kerlone), Bisoprolol/hydrochlorothiazide (Ziac), Bisoprolol (Zebeta), Carteolol (Cartrol), Metoprolol (Lopressor, Toprol XL), Nadolol (Corgard), Propranolol (Inderal), Sotalol (Betapace), Timolol (Blocadren) Beta blockers in the various classes are descπbed m the literature
PPAR gamma agonists such as thiazohdinediones (also called ghtazones) can be administered with the compounds of the disclosure in co-therapy Among the useful PPAR agonists are, for example rosightazone (Avandia), pioglitazone (Actos) and nvoghtazone Aldosterone antagonists can be administered with the compounds of the disclosure m co-therapy Among the useful Aldosterone antagonists are, for example eplerenone, spironolactone, and canrenone
Alpha blockers can be administered with the compounds of the disclosure in co-therapy Among the useful Alpha blockers are, for example Doxazosin mesylate (Cardura), Prazosin hydrochloπde (Mimpress) Prazosin and polythiazide (Mimzide), Terazosin hydrochloride (Hytrin) Alpha blockers in the various classes are descnbed in the literature
Central alpha agonists can be administered with the compounds of the disclosure in co-therapy Among the useful Central alpha agonists are, for example Clomdine hydrochloπde (Catapres), Clomdine hydrochloride and chlorthalidone (Clorpres, Combipres), Guanabenz Acetate (Wytensin), Guanfacine hydrochloπde (Tenex), Methyldopa (Aldomet), Methyldopa and chlorothiazide (Aldochlor), Methyldopa and hydrochlorothiazide (Aldoπl) Central alpha agonists in the various classes are descnbed in the literature Vasodilators can be administered with the compounds of the disclosure in co-therapy Among the useful vasodilators are, for example Isosorbide dimtrate
(Isordil), Nesiπtide (Natrecor), Hydralazine (Apresohne), Nitrates / nitroglycerin,
Minoxidil (Lomten) Vasodilators m the vaπous classes are descnbed in the literature
Blood thinners can be administered with the compounds of the disclosure in co-therapy Among the useful blood thinners are, for example Warfann (Coumadin) and Hepann Blood thinners in the vanous classes are descnbed in the literature
Anti-platelet agents can be administered with the compounds of the disclosure in co-therapy Among the useful anti-platelet agents are, for example Cyclooxygenase inhibitors (Aspiπn), Adenosine diphosphate (ADP) receptor inhibitors [Clopidogrel (Plavix), Ticlopidine (Ticlid)], Phosphodiesterase inhibitors [Cilostazol (Pletal)], Glycoprotein IIB/IIIA inhibitors [Abciximab (ReoPro), Eptifibatide (Integnlin), Tirofiban (Aggrastat), Defibrotide], Adenosine reuptake inhibitors [Dipyndamole (Persantine)] Anti-platelet agents m the vanous classes are descnbed m the literature
Lipid-lowenng agents can be administered with the compounds of the disclosure in co-therapy Among the useful lipid-lowenng agents are, for example Statins (HMG CoA reductase inhibitors), [Atorvastatm (Lipitor), Fluvastatm (Lescol), Lovastatm (Mevacor, Altoprev), Pravastatin (Pravachol), Rosuvastatm Calcium (Crestor), Simvastatin (Zocor)], Selective cholesterol absorption inhibitors [ezetimibe (Zetia)], Resms (bile acid sequestrant or bile acid-binding drugs) [Cholestyramine (Questran, Questran Light, Prevalite, Locholest, Locholest Light), Colestipol (Colestid), Colesevelam HcI (WelChol)], Fibrates (Fibπc acid derivatives) [Gemfibrozil (Lopid), Fenofibrate (Antara, Lofibra, Tπcor, and Tπghde), Clofibrate (Atromid-S)], Niacin (Nicotinic acid) Lrpid-lowermg agents m the various classes are described in the literature
The compounds of the disclosure can be used in combination with peptides or peptide analogs that activate the Guanylate Cyclase-receptor in the intestine and results in elevation of the intracellular second messenger, or cyclic guanosme monophosphate (cGMP), with increased chloride and bicarbonate secretion into the intestinal lumen and concomitant fluid secretion Example of such peptides are Linaclotide (MD-1100 Acetate), endogenous hormones guanylm and uroguanylm and enteπc bacteπal peptides of the heat stable enterotoxm family (ST peptides) and those descπbed in US 5140102, US 5489670, US 5969097, WO 2006/001931A2, WO 2008/002971A2, WO 2008/106429A2, US 2008/0227685A1 and US 7041786, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes
The compounds of the disclosure can be used in combination with type 2 chloπde channel agonists, such as Amitiza (Lubiprostone) and other related compounds descπbed in US 6414016, the entire contents of whch are incorporated herein by reference for all relevant and consistent purposes
The compounds of the disclosure can be used m combination with P2Y2 receptor agonists, such as those descπbed in EP 1196396B1 and US 6624150, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes Other agents include natnuretic peptides such as nesintide, a recombinant form of brain-natπuretic peptide (BNP) and an atπal-natπuretic peptide (ANP) Vasopressin receptor antagonists such as tolvaptan and conivaptan may be coadministered as well as phosphate binders such as renagel, renleva, phoslo and fosrenol Other agents include phosphate transport inhibitors (as descπbed in U S Pat Nos 4,806,532, 6,355,823, 6,787,528, 7,119,120, 7,109,184, U S Pat Pub No 2007/021509, 2006/0280719, 2006/0217426, International Pat Pubs WO 2001/005398, WO 2001/087294, WO 2001/082924, WO 2002/028353, WO 2003/048134, WO 2003/057225, WO2003/080630, WO 2004/085448, WO 2004/085382, European Pat Nos 1465638 and 1485391, and JP Patent No 2007131532, or phosphate transport antagonists such as Nicotinamide
2. Gastrointestinal Tract Disorders
As previously noted, the compounds described herein can be used alone or in combination with other agents For example, the compounds can be administered together with an analgesic peptide or compound The analgesic peptide or compound can be covalently attached to a compound descπbed herein or it can be a separate agent that is administered together with or sequentially with a compound descnbed herein in a combination therapy
Combination therapy can be achieved by administering two or more agents, e g , a substantially non-permeable or substantially non-bioavailable NHE- inhibiting compound descπbed herein and an analgesic peptide or compound, each of which is formulated and administered separately, or by administering two or more agents in a single formulation Other combinations are also encompassed by combination therapy For example, two agents can be formulated together and administered in conjunction with a separate formulation containing a third agent While the two or more agents in the combination therapy can be administered simultaneously, they need not be For example, administration of a first agent (or combination of agents) can precede administration of a second agent (or combination of agents) by minutes, hours, days, or weeks Thus, the two or more agents can be administered within minutes of each other or within 1, 2, 3, 6, 9, 12, 15, 18, or 24 hours of each other or within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14 days of each other or within 2, 3, 4, 5, 6, 7, 8, 9, or weeks of each other In some cases even longer intervals are possible While in many cases it is desirable that the two or more agents used in a combination therapy be present m within the patient's body at the same time, this need not be so
Combination therapy can also include two or more administrations of one or more of the agents used in the combination For example, if agent X and agent Y are used in a combination, one could administer them sequentially in any combination one or more times, e g , in the order X-Y-X, X-X-Y, Y-X-Y, Y-Y-X, X-X-Y-Y, etc
The compounds descπbed herein can be used in combination therapy with an analgesic agent, e g , an analgesic compound or an analgesic peptide The 5 analgesic agent can optionally be covalently attached to a compound descπbed herein Among the useful analgesic agents are, for example Ca channel blockers, 5HT3 agonists (e g , MCK-733), 5HT4 agonists (e g , tegaserod, prucalopπde), and 5HTl receptor antagonists, opioid receptor agonists (loperamide, fedotozine, and fentanyl), NKl receptor antagonists, CCK receptor agonists (e g , loxiglumide), NKl receptor
10 antagonists, NK3 receptor antagonists, norepmephrme-serotonm reuptake inhibitors (NSRl), vamlloid and cannabanoid receptor agonists, and sialorphm Analgesics agents in the various classes are descπbed in the literature
Opioid receptor antagonists and agonists can be administered with the compounds of the disclosure in co-therapy or linked to the compound of the disclosure,
I5 e g , by a covalent bond For example, opioid receptor antagonists such as naloxone, naltrexone, methyl nalozone, nalmefene, cypndime, beta funaltrexamme, naloxonazine, naltπndole, and nor-binaltorphimme are thought to be useful in the treatment of opioid- induced constipaption (OIC) It can be useful to formulate opioid antagonists of this type m a delayed or sustained release formulation, such that initial release of the
20 antagonist is in the mid to distal small intestine and/or ascending colon Such antagonists are descπbed in US 6,734,188 (WO 01/32180 A2), the entire contents of which are incorporated herein by reference for all relevant and consistent purposes Enkephalin pentapeptide (HOE825, Tyr-D-Lys-Gly-Phe-L-homosenne) is an agonist of the μ- and γ-opioid receptors and is thought to be useful for increasing intestinal
25 motility {Eur J Pharm , 219 445, 1992), and this peptide can be used in conjunction with the compounds of the disclosure Also useful is tπmebutme which is thought to bind to mu/delta/kappa opioid receptors and activate release of motihn and modulate the release of gastπn, vasoactive intestinal peptide, gastrin and glucagons K opioid receptor agonists such as fedotozine, ketocyclazocme, and compounds descπbed in US
30 2005/0176746 (WO 03/097051 A2), the entire contents of which are incorporated herein by reference for all relevant and consistent purposes, can be used with or linked to the compounds of the disclosure In addition, μ-opioid receptor agonists, such as morphine, diphenyloxylate, frakefamide (H-Tyr-D-Ala-Phe(F)-Phe-NH2, disclosed in WO 01/019849 Al, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes) and loperamide can be used Tyr-Arg (kyotorphm) is a dipeptide that acts by stimulating the release of met-enkephahns to elicit an analgesic effect (J Biol Chem 262 8165, 1987) Kyotorphm can be used with or linked to the compounds of the disclosure CCK receptor agonists such as caerulein from amphibians and other species are useful analgesic agents that can be used with or linked to the compounds of the disclosure Conotoxin peptides represent a large class of analgesic peptides that act at voltage gated Ca channels, NMDA receptors or nicotinic receptors These peptides can be used with or linked to the compounds of the disclosure
Peptide analogs of thymuhn (US 7,309,690 or FR 2830451, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes) can have analgesic activity and can be used with or linked to the compounds of the disclosure
CCK (CCKa or CCKb) receptor antagonists, including loxiglumide and dexloxiglumide (the R-isomer of loxiglumide) (US 5,130,474 or WO 88/05774, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes) can have analgesic activity and can be used with or linked to the compounds of the disclosure
Other useful analgesic agents include 5-HT4 agonists such as tegaserod/zelnorm and hrexapnde Such agonists are described in EP 1321142 Al, WO 03/053432A1, EP 505322 Al, EP 505322 Bl, EP 507672 Al, EP 507672 Bl, U S Pat No 5,510,353 and U S Pat No 5,273,983, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes
Calcium channel blockers such as ziconotide and related compounds descπbed in, for example, EP 625162Bl, U S Pat No 5,364,842, U S Pat No 5,587,454, U S Pat No 5,824,645, U S Pat No 5,859,186, U S Pat No 5,994,305, U S Pat No 6,087,091, U S Pat No 6,136,786, WO 93/13128 Al, EP 1336409 Al, EP 835126 Al, EP 835126 Bl, U S Pat No 5,795,864, U S Pat No 5,891,849, U S Pat No 6,054,429, WO 97/01351 Al, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes, can be used with or linked to the compounds of the disclosure
Various antagonists of the NK-I, NK-2, and NK-3 receptors (for a review see Giardma et al 2003 Drugs 6 758) can be can be used with or linked to the compounds of the disclosure
NKl receptor antagonists such as aprepitant (Merck & Co Inc), vofopitant, ezlopitant (Pfizer, Inc ), R-673 (Hoffmann-La Roche Ltd), SR-14033 and related compounds descπbed in, for example, EP 873753 Al, U S 20010006972 Al, U S 20030109417 Al, WO 01/52844 Al, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes, can be used with or linked to the compounds of the disclosure
NK-2 receptor antagonists such as nepadutant (Menaπm Ricerche SpA), saredutant (Sanofi-Synthelabo), SR-144190 (Sanofi-Synthelabo) and UK-290795 (Pfizer Inc) can be used with or linked to the compounds of the disclosure
NK3 receptor antagonists such as osanetant (Sanofi-Synthelabo), talnetant and related compounds descnbed in, for example, WO 02/094187 A2, EP 876347 Al, WO 97/21680 Al, U S Pat No 6,277,862, WO 98/11090, WO 95/28418, WO 97/19927, and Boden et al (J Med Chem 39 1664-75, 1996) , the entire contents of which are incorporated herein by reference for all relevant and consistent purposes, can be used with or linked to the compounds of the disclosure
Norepinephrme-serotonin reuptake inhibitors such as milnacipran and related compounds described in WO 03/077897 Al, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes, can be used with or linked to the compounds of the disclosure
Vamlloid receptor antagonists such as arvaml and related compounds descπbed m WO 01/64212 Al, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes, can be used with or linked to the compounds of the disclosure The compounds can be used in combination therapy with a phosphodiesterase inhibitor (examples of such inhibitors can be found in U S Pat No 6,333,354, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes)
The compounds can be used alone or in combination therapy to treat disorders associated with chloride or bicarbonate secretion that may lead to constipation, e g , Cystic Fibrosis
The compounds can also or alternatively be used alone or in combination therapy to treat calcium-mduced constipation effects Constipation is commonly found in the geriatric population, particularly patients with osteoporosis who have to take calcium supplements Calcium supplements have shown to be beneficial in ostoporotic patients to restore bone density but compliance is poor because of constipation effects associated therewith
The compounds of the current disclosure have can be used in combination with an opioid Opioid use is mainly directed to pam relief, with a notable side-effect being GI disorder, e g constipation These agents work by binding to opioid receptors, which are found principally in the central nervous system and the gastrointestinal tract The receptors in these two organ systems mediate both the beneficial effects, and the undesirable side effects (e g decrease of gut motility and ensuing constipation) Opioids suitable for use typically belong to one of the following exemplary classes natural opiates, alkaloids contained in the resin of the opium poppy including morphine, codeine and thebaine, semi-synthetic opiates, created from the natural opioids, such as hydromorphone, hydrocodone, oxycodone, oxymorphone, desomorphme, diacetylmorphine (Heroin), mcomorphme, dipropanoylmorphine, benzylmorphine and ethylmorphine, fully synthetic opioids, such as fentanyl, pethidine, methadone, tramadol and propoxyphene, endogenous opioid peptides, produced naturally in the body, such as endorphins, enkephalins, dynorphms, and endomorphins
The compound of the disclosure can be used alone or in combination therapy to alleviate GI disorders encountered with patients with renal failure (stage 3- 5) Constipation is the second most reported symptom in that category of patients (Murtagh et al , 2006, Murtagh et al , 2007a, Murtagh et al , 2007b) Without being held by theory, it is believed that kidney failure is accompanied by a stimulation of intestinal Na re-absorption (Hatch and Freel, 2008) A total or partial inhibition of such transport by administration of the compounds of the disclosure can have a therapeutic benefit to improve GI transit and relieve abdominal pain In that context, the compounds of the disclosure can be used in combination with Angiotensin-modulating agents Angiotensin Converting Enzyme (ACE) inhibitors (e g captopπl, enalopπl, 5 hsmopπl, ramipπl) and Angiotensin II receptor antagonist therapy (also referred to as ATi-antagonists or angiotensin receptor blockers, or ARB's), diuretics such as loop diuretics (e g furosemide, bumetamde), Thiazide diuretics (e g hydrochlorothiazide, chlorthalidone, chlorthiazide) and potassium-spaπng diuretics amilonde, beta blockers bisoprolol, carvedilol, nebivolol and extended-release metoprolol, positive inotropes
10 Digoxin, dobutamme, phosphodiesterase inhibitors such as milrinone, alternative vasodilators combination of isosorbide dimtrate/hydralazine, aldosterone receptor antagonists spironolactone, eplerenone, natnuretic peptides Nesiπtide, a recombinant form of bram-natπuretic peptide (BNP), atπal-natπuretic peptide (ANP), vasopressin receptor antagonists Tolvaptan and comvaptan, phosphate binder (Renagel, Renleva,
15 Phoslo, Fosrenol), phosphate transport inhibitor such as those descπbed in US 4806532, US 6355823, US 6787528, WO 2001/005398, WO 2001/087294, WO 2001/082924, WO 2002/028353, WO 2003/048134, WO 2003/057225, US 7119120, EP 1465638, US Appl 2007/021509, WO 2003/080630, US 7109184, US Appl 2006/0280719 , EP 1485391, WO 2004/085448, WO 2004/085382, US Appl 2006/0217426, JP
2Q 2007/131532, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes, or phosphate transport antagonist (Nicotinamide)
The compounds of the disclosure can be used in combination with peptides or peptide analogs that activate the Guanylate Cyclase-receptor in the intestine and results in elevation of the intracellular second messenger, or cyclic guanosine
25 monophosphate (cGMP), with increased chloride and bicarbonate secretion into the intestinal lumen and concomitant fluid secretion Example of such peptides are Lmaclotide (MD-1100 Acetate), endogenous hormones guanyhn and uroguanylm and enteric bacteπal peptides of the heat stable enterotoxm family (ST peptides) and those descπbed in US 5140102, US 5489670, US 5969097, WO 2006/001931A2, WO
30 2008/002971A2, WO 2008/106429A2, US 2008/0227685A1 and US 7041786, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes
The compounds of the disclosure can be used in combination with type-2 chloπde channel agonists, such as Amitiza (Lubiprostone) and other related compounds described in US 6414016, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes
The compounds of the disclosure can be used in combination with P2Y2 receptor agonists, such as those descπbed in EP 1196396B1 and US 6624150, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes
The compounds of the disclosure can be used in combination with laxative agents such as bulk-producing agents, e g psyllium husk (Metamucil), methylcellulose (Citrucel), polycarbophil, dietary fiber, apples, stool softeners/surfactant such as docusate (Colace, Diocto), hydrating agents (osmotics), such as dibasic sodium phosphate, magnesium citrate, magnesium hydroxide (Milk of magnesia), magnesium sulfate (which is Epsom salt), monobasic sodium phosphate, sodium biphosphate, hyperosmotic agents glycenn suppositones, sorbitol, lactulose, and polyethylene glycol (PEG) The compounds of the disclosure can be also be used in combination with agents that stimulate gut peristalsis, such as Bisacodyl tablets (Dulcolax), Casanthranol, Senna and Alom, from Aloe Vera
In one embodiment, the compounds of the disclosure accelerate gastrointestinal transit, and more specifically in the colon, without substantially affecting the residence time in the stomach, i e with no significant effect on the gastπc emptying time Even more specifically the compounds of the invention restore colonic transit without the side-effects associated with delayed gastric emptying time, such as nausea The GI and colonic transit are measured in patients using methods reported in, for example Burton DD, Camilleπ M, Mullan BP, et al , J Nucl Med , 1997,38 1807- 1810, Cremonini F, Mullan BP, Camilleπ M, et al , Aliment Pharmacol Ther , 2002,16 1781-1790, Camilleπ M, Zinsmeister AR, Gastroenterology, 1992,103 36-42, Bouras EP, Camilleπ M, Burton DD, et al , Gastroenterology, 2001,120 354-360, Coulie B, Szarka LA, Camilleπ M, et al , Gastroenterology, 2000,119 41-50, Prather CM, Camilleπ M, Zmsmeister AR, et al , Gastroenterology, 2000,118 463^68, and, Camilleπ M, McKjnzie S, Fox J, et al , Clin Gastroenterol Hepatol , 2004,2 895-904
C. Polymer Combination Therapy The NHE-inhibiting compounds descπbed therein may be administered to patients in need thereof in combination with a fluid-absorbing polymer ("FAP") The intestinal fluid-absorbing polymers useful for administration in accordance with embodiments of the present disclosure may be administered orally in combination with non-absorbable NHE-inhibitors (e g , a NHE-3 inhibitor) to absorb the intestinal fluid resulting from the action of the sodium transport inhibitors Such polymers swell in the colon and bind fluid to impart a consistency to stools that is acceptable for patients The fluid-absorbing polymers descπbed herein may be selected from polymers with laxative properties, also referred to as bulking agents (i e , polymers that retain some of the intestinal fluid m the stools and impart a higher degree of hydration in the stools and facilitate transit) The flmd-absorbmg polymers may also be optionally selected from pharmaceutical polymers with anti-diarrhea function, i e , agents that maintain some consistency to the stools to avoid watery stools and potential incontinence
The ability of the polymer to maintain a certain consistency in stools with a high content of fluid can be characterized by its "water holding power " Wenzl et al (in Determinants of decreased fecal consistency in patients with diarrhea, Gastroenterology, v 108, no 6, p 1729-1738 (1995)) studied the determinants that control the consistency of stools of patients with diarrhea and found that they were narrowly correlated with the water holding power of the feces The water holding power is determined as the water content of given stools to achieve a certain level of consistency (corresponding to "formed stool" consistency) after the reconstituted fecal matter has been centπfuged at a certain g number Without being held to any particular theory, has been found that the water holding power of the feces is increased by ingestion of certain polymers with a given fluid absorbing profile More specifically, it has been found that the water-holding power of said polymers is correlated with their fluid absorbancy under load (AUL), even more specifically the AUL of said polymers is greater than 15 g of isotonic flmd/g of polymer under a static pressure of 5kPa, even more preferably under a static pressure of 1OkPa
The FAP utilized in the treatment method of the present disclosure preferably has a AUL of at least about 1O g, about 15 g, about 2O g, aboug 25 g or more of isotonic fluid/g of polymer under a static pressure of about 5 kPa, and preferably about 10 kPA, and may have a fluid absorbency of about 20 g, about 25 g or more, as determined using means generally known in the art Additionally or alternatively, the FAP may impart a minimum consistency to fecal matter and, in some embodiments, a consistency graded as "soft" in the scale described in the test method below, when fecal non water-soluble solid fraction is from 10% to 20%, and the polymer concentration is from 1% to 5% of the weight of stool The determination of the fecal non water-soluble solid fraction of stools is described in Wenz et al The polymer may be uncharged or may have a low charge density (e g , 1-2 meq/gr) Alternatively or m addition, the polymer may be delivered directly to the colon using known delivery methods to avoid premature swelling m the esophagus
In one embodiment of the present disclosure, the FAP is a "superabsorbent" polymer (i e , a lightly crosslinked, partially neutralized polyelectrolyte hydrogel similar to those used in baby diapers, feminine hygiene products, agriculture additives, etc ) Superabsorbent polymers may be made of a lightly crosslinked polyacrylate hydrogel The swelling of the polymer is dnven essentially by two effects (i) the hydration of the polymer backbone and entropy of mixing and (ii) the osmotic pressure arising from the counter-ions (e g , Na ions) within the gel The gel swelling ratio at equilibrium is controlled by the elastic resistance inherent to the polymer network and by the chemical potential of the bathing fluid, i e , the gel will de-swell at higher salt concentration because the background electrolyte will reduce the apparent charge density on the polymer and will reduce the difference of free ion concentrations inside and outside the gel that dπves osmotic pressure The swelling ratio SR (g of fluid per g of dry polymer and synonymously "fluid absorbency") may vary from 1000 in pure water down to 30 m 0 9% NaCl solution representative of physiological saline (i e , isotonic) SR may increase with the degree of neutralization and may decrease with the crosslmkmg density SR generally decreases with an applied load with the extent of reduction dependent on the strength of the gel, i e , the crosslinking density The salt concentration withm the gel, as compared with the external solution, may be lower as a result of the Donnan effect due to the internal electrical potential The fluid-absorbing polymer may include crosslinked polyacrylates which are fluid absorbent such as those prepared from α,β-ethylemcally unsaturated monomers, such as monocarboxylic acids, polycarboxylic acids, acrylamide and their deπvatives These polymers may have repeating units of acrylic acid, methacrylic acid, metal salts of acrylic acid, acrylamide, and acrylamide deπvatives (such as 2- acrylamido-2-methylpropanesulfomc acid) along with vaπous combinations of such repeating units as copolymers Such denvatives include acrylic polymers which include hydrophilic grafts of polymers such as polyvinyl alcohol Examples of suitable polymers and processes, including gel polymerization processes, for preparing such polymers are disclosed in U S Pat Nos 3,997,484, 3,926,891, 3,935,099, 4,090,013, 4,093,776, 4,340,706, 4,446,261 , 4,683,274, 4,459,396, 4,708,997, 4,076,663, 4,190,562, 4,286,082, 4,857,610, 4,985,518, 5,145,906, 5,629,377 and 6,908,609 which are incorporated herein by reference for all relevant and consistent purposes (in addition, see Buchholz, F L and Graham, A T , "Modern Superabsorbent Polymer Technology," John Wiley & Sons (1998), which is also incorporated herein by reference for all relevant and consistent purposes) A class of preferred polymers for treatment in combination with NHE-inhibitors is polyelectrolytes
The degree of crosslinking can vary greatly depending upon the specific polymer material, however, m most applications the subject superabsorbent polymers are only lightly crosslinked, that is, the degree of crosslinking is such that the polymer can still absorb over 10 times its weight in physiological saline (i e , 0 9% saline) For example, such polymers typically include less than about 02 mole % crosslinking agent
In some embodiments, the FAP's utilized for treatment are Calcium Carbophil (Registry Number 9003-97-8, also referred as Carbopol EX-83), and Carpopol 934P In some embodiments, the fluid-absorbmg polymer is prepared by high internal phase emulsion ("HIPE") processes The HIPE process leads to polymeric foam slabs with a very large porous fraction of interconnected large voids (about 100 microns) (i e , open-cell structures) This technique produces flexible and collapsible foam mateπals with exceptional suction pressure and fluid absorbency (see U S Patent Nos 5,650,222, 5,763,499 and 6,107,356, which are incorporated herein for all relevant and consistent purposes) The polymer is hydrophobic and, therefore, the surface should be modified so as to be wetted by the aqueous fluid This is accomplished by post-treating the foam mateπal by a surfactant in order to reduce the interfacial tension These materials are claimed to be less compliant to loads, i e , less prone to de-swellmg under static pressure
In some embodiments, fluid-absorbmg gels are prepared by aqueous free radical polymerization of acrylamide or a deπvative thereof, a crosslinker (e g , methylene-bis-acrylamide) and a free radical initiator redox system in water The mateπal is obtained as a slab Typically the swelling ratio of crosslmked polyacrylamide at low crosslinking density (e g , 2%-4% expressed as weight % of methylene-bis-acrylamide) is between 25 and 40 (F Horkay, Macromolecules, 22, pp 2007-09 (1989)) The swelling properties of these polymers have been extensively studied and are essentially the same of those of crosslmked polyacrylic acids at high salt concentration Under those conditions, the osmotic pressure is null due to the presence of counter-ions and the swelling is controlled by the free energy of mixing and the network elastic energy Stated differently, a crosslmked polyacrylamide gel of same crosslink density as a neutralized polyacrylic acid will exhibit the same swelling ratio (i e , fluid absorbing properties) and it is believed the same degree of deswellmg under pressure, as the crosslmked polyelectrolyte at high salt content (e g , 1 M) The properties (e g , swelling) of neutral hydrogels will not be sensitive to the salt environment as long as the polymer remains in good solvent conditions Without being held to any particular theory, it is believed that the fluid contained within the gel has the same salt composition than the surrounding fluid (i e , there is no salt partitioning due to Donnan effect) Another subclass of fluid-absorbmg polymers that may be utilized is hydrogel mateπals that include N-alkyl acrylamide polymers (e g , N- isopropylacrylamide (NIPAM)) The corresponding aqueous polyNIPAM hydrogel shows a temperature transition at about 35°C Above this temperature the hydrogel may collapse The mechanism is generally reversible and the gel re-swells to its original swelling ratio when the temperature reverts to room temperature This allows production of nanoparticles by emulsion polymerization (R Pelton, Advances in Colloid and Interface Science, 85, pp 1-33, (2000)) The swelling characteπstics of poly-NIPAM nanoparticles below the transition temperature have been reported and are similar to those reported for bulk gel of polyNIPAM and equivalent to those found for polyacrylamide (i e 30-50 g/g) (W McPhee, Journal of Colloid and Interface Science, 156, pp 24-30 (1993), and, K Oh, Journal of Applied Polymer Science 69, pp 109- 114 (1997))
In some embodiments, the FAP utilized for treatment in combination with a NHE-mhibitor is a superporous gel that may delay the emptying of the stomach for the treatment of obesity (J Chen, Journal of Controlled Release, 65, pp 73-82 (2000), or to deliver proteins Polyacrylate-based SAP's with a macroporous structure may also be used Macroporous SAP and superporous gels differ in that the porous structure remains almost intact in the dry state for superporous gels, but disappears upon drying for macroporous SAP's The method of preparation is different although both methods use a foaming agent (e g , carbonate salt that generates CO2 bubbles duπng polymerization) Typical swelling ratios, SR, of superporous matenals are around 10 Superporous gels keep a large internal pore volume in the dry state
Macroporous hydrogels may also be formed using a method whereby polymer phase separation in induced by a non-solvent The polymer may be polyNIPAM and the non-solvent utilized may be glucose (see, e g , Z Zhang, J Org Chem , 69, 23 (2004)) or NaCl (see, e g , Cheng et al , Journal of Biomedical Matenals Research - Part A, VoI 67, Issue 1, 1 October 2003, Pages 96-103) The phase separation induced by the presence of NaCl leads to an increase in swelling ratio These matenals are preferred if the swelling ratio of the material, SR, is maintained in salt isotonic solution and if the gels do not collapse under load The temperature of "service" should be shifted beyond body temperature, e g by diluting NIPAM in the polymer with monomer devoid of transition temperature phenomenon
In some embodiments, the fluid-absorbing polymer may be selected from certain naturally-occurring polymers such as those containing carbohydrate moieties In a preferred embodiment, such carbohydrate-containing hydrogels are non- digestible, have a low fraction of soluble mateπal and a high fraction of gel-formmg materials In some embodiments, the fluid-absorbmg polymer is selected from xanthan, guar, wellan, hemicelluloses, alkyl-cellulose, hydro-alkyl-cellulose, carboxy-alkyl- cellulose, carrageenan, dextran, hyaluronic acid and agarose In a preferred embodiment, the gel forming polymer is psyllium Psyllium (or "lspaghula") is the common name used for several members of the plant genus Plantago whose seeds are used commercially for the production of mucilage Most preferably, the fluid-absorbing polymer is in the gel-formmg fraction of psyllium, i e , a neutral sacchaπde copolymer of arabmose (25%) and xylose (75%) as characterized in (J Marlett, Proceedings of the Nutrition Society, 62, pp 2-7-209 (2003), and, M Fischer, Carbohydrate Research, 339, 2009-2012 (2004)), and further described in U S Pat Nos 6,287,609, 7,026,303, 5,126,150, 5,445,831, 7,014,862, 4,766,004, 4,999,200, each of which is incorporated herein for all relevant and consistent purposes, and over-the-counter psillium-contaimng agents such as those marketed under the name Metamucil (The Procter and Gamble company) Preferably the a psyllium-contaimng dosage form is suitable for chewing, where the chewing action disintegrates the tablet into smaller, discrete particles pnor to swallowing but which undergoes minimal gelling m the mouth, and has acceptable mouthfeel and good aesthetics as perceived by the patient
The psyllium-contaimng dosage form includes physically discrete unit suitable as a unitary dosage for human subjects and other mammals, each containing a predetermined quantity of active mateπal (e g the gel-forming polysaccharide) calculated to produce the desired therapeutic effect Solid oral dosage forms that are suitable for the present compositions include tablets, pills, capsules, lozenges, chewable tablets, troches, cachets, pellets, wafer and the like In some embodiments, the FAP is a polysaccharide particle wherein the polysacchaπde component includes xylose and arabmose The ratio of the xylose to the arabinose may be at least about 3 1 by weight, as described in U S Pat Nos 6,287,609, 7,026,303 and 7,014,862, each of which is incorporated herein for all relevant and consistent purposes
The fluid-absorbing polymers descπbed herein may be used in combination with the NHE-mhibiting compounds or a pharmaceutical composition containing the compound The NHE inhibitor and the FAP may also be administered with other agents including those descπbed under the heading "Combination Therapies" without departing from the scope of the present disclosure As descπbed above, the NHE inhibitor may be administered alone without use of a fluid-absorbing polymer to resolve symptoms without eliciting significant diarrhea or fecal fluid secretion that would require the co-admimstration of a fluid-absorbing polymer
The fluid-absorbing polymers descπbed herein may be selected so as to not induce any substantial interaction with the NHE-inhibitmg compounds or a pharmaceutical composition containing the compound As used herein, "no substantial interaction" generally means that the co-admimstration of the FAP polymer would not substantially alter (i e , neither substantially decrease nor substantially increase) the pharmacological property of the NHE-inhibiting compounds administered alone For example, FAPs containing negatively charged functionality, such as carboxylates, sulfonates, and the like, may potentially interact ionically with positively charged NHE inhibitors, preventing the inhibitor from reaching its pharmacological target In addition, it may be possible that the shape and arrangement of functionality in a FAP could act as a molecular recognition element, and sequestor NHE inhibitors via "host- guest" interactions via the recognition of specific hydrogen bonds and/or hydrophobic regions of a given inhibitor Accordingly, in vanous embodiments of the present disclosure, the FAP polymer may be selected, for co-admimstration or use with a compound of the present disclosure, to ensure that (i) it does not ionically interact with or bind with the compound of the present disclosure (by means of, for example, a moiety present therein possessing a charge opposite that of a moiety in the compound itself), and/or (ii) it does not possess a charge and/or structural conformation (or shape or arrangement) that enables it to establish a "host-guest" interaction with the compound of the present disclosure (by means of, for example, a moiety present therein that may act as a molecular recognition element and sequester the NHE inhibitor or inhibiting moiety of the compound)
D. Dosage It is to be noted that, as used herein, an "effective amount" (or
"pharmaceutically effective amount") of a compound disclosed herein, is a quantity that results m a beneficial clinical outcome of the condition being treated with the compound compared with the absence of treatment The amount of the compound or compounds administered will depend on the degree, seventy, and type of the disease or condition, the amount of therapy desired, and the release characteπstics of the pharmaceutical formulation It will also depend on the subject's health, size, weight, age, sex and tolerance to drugs Typically, the compound is administered for a sufficient peπod of time to achieve the desired therapeutic effect
In embodiments wherein both an NHE-inhibitor compound and a fluid- absorbing polymer are used in the treatment protocol, the NHE-inhibitor and FAP may be administered together or in a "dual-regimen" wherein the two therapeutics are dosed and administered separately When the NHE inhibitor and the fluid-absorbing polymer are dosed separately, the typical dosage administered to the subject in need of the NHE inhibitor is typically from about 5 mg per day and about 5000 mg per day and, in other embodiments, from about 50 mg per day and about 1000 mg per day Such dosages may induce fecal excretion of sodium (and its accompanying anions), from about 10 mmol up to about 250 mmol per day, from about 20 mmol to about 70 mmol per day or even from about 30 mmol to about 60 mmol per day
The typical dose of the fluid-absorbing polymer is a function of the extent of fecal secretion induced by the non-absorbable NHE inhibitor Typically the dose is adjusted according to the frequency of bowel movements and consistency of the stools More specifically the dose is adjusted so as to avoid liquid stools and maintain stool consistency as "soft" or semi-formed, or formed To achieve the desired stool consistency and provide abdominal relief to patients, typical dosage ranges of the fluid- absorbing polymer to be administered in combination with the NHE inhibitor, are from about 2 g to about 50 g per day, from about 5 g to about 25 g per day or even from about 10 g to about 20 g per day When the NHE-mhibitor and the FAP are administered as a single dosage regimen, the daily uptake may be from about 2 g to about 50 g per day, from about 5 g to about 25 g per day, or from about 10 g to about 20 g per day, with a weight ratio of NHE inhibitor to fluid- absorbing polymer being from 5 about 1 1000 to 1 10 or even from about 1 500 to 1 5 or about 1 100 to 1 5
A typical dosage of the substantially impermeable or substantially systemically non-bioavailable, NHE-mhibiting compound when used alone without a FAP may be between about 0 2 mg per day and about 2 g per day, or between about 1 mg and about 1 g per day, or between about 5 mg and about 500 mg, or between about
10 10 mg and about 250 mg per day, which is administered to a subject in need of treatment
The frequency of administration of therapeutics descπbed herein may vary from once-a-day (QD) to twice-a-day (BID) or fhπce-a-day (TID), etc , the precise frequency of administration varying with, for example, the patient's condition, the
15 dosage, etc For example, in the case of a dual -regimen, the NHE-mhibitor could be taken once-a-day while the fluid-absorbmg polymer could be taken at each meal (TID)
E. Modes of Administration
The substantially impermeable or substantially systemically non- 20 bioavailable, NHE-inhibiting compounds of the present disclosure with or without the fluid-absorbing polymers descπbed herein may be administered by any suitable route The compound is preferably administrated orally (e g , dietary) in capsules, suspensions, tablets, pills, dragees, liquids, gels, syrups, slurries, and the like Methods for encapsulating compositions (such as m a coating of hard gelatin or cyclodextran) are 25 known in the art (Baker, et al , "Controlled Release of Biological Active Agents", John Wiley and Sons, 1986) The compounds can be administered to the subject in conjunction with an acceptable pharmaceutical earner as part of a pharmaceutical composition The formulation of the pharmaceutical composition will vary according to the route of administration selected Suitable pharmaceutical earners may contain 30 inert ingredients which do not interact with the compound The earners are biocompatible, i e , non-toxic, non-mflammatory, non-immunogenic and devoid of other undesired reactions at the administration site. Examples of pharmaceutically acceptable earners include, for example, saline, commercially available inert gels, or liquids supplemented with albumin, methyl cellulose or a collagen matrix Standard pharmaceutical formulation techniques can be employed, such as those described in 5 Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa
Pharmaceutical preparations for oral use can be obtained by combining a compound of the present disclosure with a solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in
10 particular, fillers such as sugars, including lactose, sucrose, manmtol, or sorbitol, cellulose preparations such as, for example, maize starch, wheat starch, πce starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP) If desired, disintegrating agents can be added, such as cross-linked polyvinyl pyrrolidone, agar, or
I5 algimc acid or a salt thereof such as sodium alginate
Dragee cores are provided with suitable coatings For this purpose, concentrated sugar solutions can be used, which can optionally contain gum arable, talc, polyvinyl pyrrohdone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures Dyestuffs or
2Q pigments can be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses
Pharmaceutical preparations which can be used orally include push-fit capsules made of a suitable mateπal, such as gelatin, as well as soft, sealed capsules made of a suitable mateπal, for example, gelatin, and a plasticizer, such as glycerol or
25 sorbitol The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubπcants such as talc or magnesium stearate and, optionally, stabilizers In soft capsules, the active compounds can be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols In addition, stabilizers can be added All formulations for
30 oral administration should be in dosages suitable for such administration It will be understood that, certain compounds of the disclosure may be obtained as different stereoisomers (e g , diastereomers and enantiomers) or as isotopes and that the disclosure includes all isomenc forms, racemic mixtures and isotopes of the disclosed compounds and a method of treating a subject with both pure isomers and mixtures thereof, including racemic mixtures, as well as isotopes Stereoisomers can be separated and isolated using any suitable method, such as chromatography
F. Delayed Release
NHE proteins show considerable diversity in their patterns of tissue expression, membrane localization and functional roles (See, e g , The sodium- hydrogen exchanger - From molecule To Its Role In Disease, Karmazyn, M , Avkiran, M , and Fhegel, L , eds , Kluwer Academics (2003) )
In mammals, nine distinct NHE genes (NHE-I through -9) have been descnbed Of these nine, five (NHE-I through -5) are principally active at the plasma membrane, whereas NHE-6, -7 and -9 reside predominantly within intracellular compartments
NHE-I is ubiquitously expressed and is chiefly responsible for restoration of steady state intracellular pH following cytosolic acidification and for maintenance of cell volume Recent findings show that NHE-I is crucial for organ function and survival (e g NHE-I -null mice exhibit locomotor abnormalities, epileptic- hke seizures and considerable mortality before weaning)
In contrast with NHE-I expressed at the basolateral side of the nephrons and gut epithelial cells, NHE-2 through -4 are predominantly expressed on the apical side of epitheha of the kidney and the gastrointestinal tract Several lines of evidence show that NHE-3 is the major contπbutor of renal bulk Na+ and fluid re-absorption by the proximal tubule The associated secretion of H+ by NHE-3 into the lumen of renal tubules is also essential for about 2/3 of renal HCO3 re-absorption Complete disruption of NHE-3 function in mice causes a sharp reduction in HCO3 , Na+ and fluid re-absorption in the kidney, which is consistently associated with hypovolemia and acidosis In one embodiment, the novel compounds of the invention are intended to target the apical NHE antiporters (e g NHE-3, NHE-2 and NHE-8) without substantial permeability across the layer of gut epithelial cells, and/or without substantial activity towards NHEs that do not reside predominantly in the GI tract This invention provides a method to selectively inhibit GI apical NHE antiporters and provide the desired effect of salt and fluid absorption inhibition to correct abnormal fluid homeostasis leading to constipations states Because of their absence of systemic exposure, said compounds do not interfere with other key physiological roles of NHEs highlighted above For instance, the compounds of the invention are expected to treat constipation in patients in need thereof, without eliciting undesired systemic effects, such as for example salt wasting or bicarbonate loss leading to hyponatremia and acidosis among other disorders
In another embodiment, the compounds of the invention are delivered to the small bowel with little or no interaction with the upper GI such as the gastric compartment and the duodenum The applicant found that an early release of the compounds in the stomach or the duodenum can have an untoward effect on gastπc secretion or bicarbonate secretion (also referred to as "bicarbonate dump") In this embodiment the compounds are designed so as to be released in an active form past the duodenum This can be accomplished by either a prodrug approach or by specific drug delivery systems
As used herein, "prodrug" is to be understood to refer to a modified form of the compounds detailed herein that is inactive (or significantly less active) in the upper GI, but once administered is metabolised in vivo into an active metabolite after getting past, for example, the duodenum Thus, in a prodrug approach, the activity of the NHE inhibitor can be masked with a transient protecting group that is liberated after compound passage through the desired gastπc compartment For example, acylation or alkylation of the essential guanidinyl functionality of the NHE inhibitor would render it biochemically inactive, however, cleavage of these functional groups by intestinal amidases, esterases, phosphatases , and the like, as well enzymes present in the colonic flora, would liberate the active parent compound Prodrugs can be designed to exploit the relative expression and localization of such phase I metabolic enzymes by carefully optimizing the structure of the prodrug for recognition by specific enzymes As an example, the antiinflammatory agent sulfasalazine is converted to 5-aminosahcylate in the colon by reduction of the diazo bond by intestinal bacteria
In a drug delivery approach the NHE-inhibitor compounds of the invention are formulated in certain pharmaceutical compositions for oral administration that release the active in the targeted areas of the GI, i e , jejunum, ileum or colon, or preferably the distal ileum and colon, or even more preferably the colon
Methods known from the skilled-in-the art are applicable (See, e g , Kumar, P and Mishra, B , Colon Targeted Drug Delivery Systems - An Overview, Curr Drug Deliv 2008, J (3), 186-198, Jam, S K and Jam, A , Target-specific Drug Release to the Colon , Expert Opin Drug Dehv 2008, 5 (5), 483-498, Yang, L , Biorelevant Dissolution Testing of Colon-Specific Delivery Systems Activated by Colonic Microflora, J Control Release 2008, 125 (2), 77-86, Siepmann, F , Siepmann, J , Walther, M , MacRae, R J , and Bodmeier, R , Polymer Blends for Controlled Release Coatings, J Control Release 2008, 125 (1), 1 15, Patel, M , Shah, T , and Amin, A , Therapeutic Opportunities in Colon-Specific Drug-Delivery Systems, Crit Rev Ther Drug Carrier Syst , 2007, 24 (2), 147-202, Jain, A , Gupta, Y , Jain, S K , Perspectives of Biodegradable Natural Polysaccharides for Site-specific Drug Delivery to the Colon , J Pharm Sa 2007, 10 (1), 86 128, Van den, M G , Colon Drug Delivery, Expert Opin Drug Dehv , 2006, 3 (1), 111-125, Basit, A W , Advances in Colonic Drug Delivery, Drugs 2005, 65 (14), 1991-2007, Chourasia, M K , Jam, S K , Polysaccharides for Colon-Targeted Drug Delivery, Drug Dehv 2004, ;; (2), 129-148, Shareef, M A , Khar, R K , Ahuja, A , Ahmad, F J , and Raghava, S , Colonic Drug Delivery An Updated Review, AAPS Pharm Sa 2003, 5 (2), El 7, Chourasia, M K , Jain, S K , Pharmaceutical Approaches to Colon Targeted Drug Delivery Systems, J Pharm Sci 2003, 6 (1), 33 66, and, Sinha, V R , Kumna, R , Colonic Drug Delivery Prodrug Approach, Pharm Res 2001, 18 (5), 557-564 Typically the active pharmaceutical ingredient (API) is contained in a tablet / capsule designed to release said API as a function of the environment (e g , pH, enzymatic activity, temperature, etc ), or as a function of time One example of this approach is Eudracol™ (Pharma Polymers Business Line of Degussa's Specialty Acrylics Business Unit), where the API-contaimng core tablet is layered with various polymeric coatings with specific dissolution profiles The first layer ensures that the tablet passes through the stomach mtact so it can continue through the small intestine The change from an acidic environment in the stomach to an alkaline environment in the small intestine initiates the release of the protective outer layer As it travels through the colon, the next layer is made permeable by the alkalinity and intestinal fluid This allows fluid to penetrate to the intenor layer and release the active ingredient, which diffuses from the core to the outside, where it can be absorbed by the intestinal wall Other methods are contemplated without departing from the scope of the present disclosure In another example, the pharmaceutical compositions of the invention can be used with drug earners including pectin and galactomannan, polysacchaπdes that are both degradable by colonic bacteπal enzymes (See, e g , U S Pat No 6,413,494, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes ) While pectin or galactomannan, if used alone as a drug earner, are easily dissolved m simulated gastric fluid and simulated intestinal fluid, a mixture of these two polysacchandes prepared at a pH of about 7 or above produces a strong, elastic, and insoluble gel that is not dissolved or disintegrated m the simulated gastnc and intestinal fluids, thus protecting drugs coated with the mixture from being released in the upper GI tract When the mixture of pectm and galactomannan arnves in the colon, it is rapidly degraded by the synergic action of colonic bactenal enzymes In yet another aspect, the compositions of the invention may be used with the pharmaceutical matnx of a complex of gelatin and an anionic polysacchande (e g , pectinate, pectate, alginate, chondroitm sulfate, polygalacturonic acid, tragacanth gum, arable gum, and a mixture thereof), which is degradable by colonic enzymes (U S Pat No 6,319,518)
In yet other embodiments, fluid-absorbing polymers that are administered m accordance with treatment methods of the present disclosure are formulated to provide acceptable/pleasant organoleptic properties such as mouthfeel, taste, and/or to avoid premature swelling/gelation in the mouth and in the esophagus and provoke choking or obstruction The formulation may be designed in such a way so as to ensure the full hydration and swelling of the FAP in the GI tract and avoid the formation of lumps. The oral dosages for the FAP may take various forms including, for example, powder, granulates, tablets, wafer, cookie and the like, and are most preferably delivered to the small bowel with little or no interaction with the upper GI such as the gastric compartment and the duodenum .
S The above-described approaches or methods are only some of the many methods reported to selectively deliver an active in the lower part of the intestine, and therefore should not be viewed to restrain or limit the scope of the disclosure.
The following non-limiting examples are provided to further illustrate Q the present disclosure.
EXAMPLES
Exemplary Compound Synthesis
Example 1
2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethylphosphonic acid
Figure imgf000146_0001
Intermediate 1.1: 2-bromo-l-(3-bromophenyl)ethanone: Into a 500-mL 3-necked round-bottom flask, was placed a solution of l-(3-bromophenyl)ethanone (40 g, 202 02 mmol, 1 00 eqmv) in acetic acid (200 mL) This was followed by the addition of a solution of Br2 (32 g, 200 00 mmol) in acetic acid (50 mL) dropwise with stimng at 6O0C The resulting solution was stirred for 3 h at 6O0C m an oil bath The resulting mixture was concentrated under vacuum The crude product was re-crystallized from petroleum ether ethyl acetate in the ratio of 8 1 This resulted in 24 g (43%) of 2- bromo-l-(3-bromophenyl)ethanone as a yellow solid
Figure imgf000146_0002
Intermediate 1.2: l-(3-bromophenyl)-2-((2,4- dichlorobenzyl)(methyl)amino)ethanone: Into a IL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 2- bromo l-(3-bromophenyl)ethanone (55 g, 199 28 mmol, 1 00 equiv) m 1,4-dioxane (300 mL), TEA (40 g, 396 04 mmol, 1 99 eqmv), and (2,4-dichlorophenyl)-N- methylmethanamme (38 g, 201 06 ininol, 1 01 eqmv) The resulting solution was stirred for 2 h at 250C m an oil bath The solids were filtered out and the filtrate was used without any further purification
Figure imgf000147_0001
Intermediate 1.3: l-(3-bromophenyl)-2-((2,4- dichlorobenzyl)(methyl)amino)ethano]: Into a IL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 2- ((2,4-dichlorobenzyl)(methyl)ammo)-l-(3-bromophenyl)ethanone (77 g, 198 97 mmol, 1 00 eqmv, theoretical yield) in methanol (300 mL) This was followed by the addition of NaBH4 (15 g, 394 74 mmol, 1 98 equiv) in several batches at O0C The resulting solution was stirred for 30 min at O0C m a water/ice bath The reaction was then quenched by the addition of 100 mL of acetone The resulting mixture was concentrated under vacuum The resulting solution was extracted with 3x100 mL of ethyl acetate and the organic layers combined and dπed over anhydrous sodium sulfate The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 100) This resulted in 50 g (65%) of 2-((2,4-dichlorobenzyi)(methyi)amino)-l-(3- bromophenyl)ethanol as a yellow oil
Figure imgf000147_0002
Intermediate 1.4: 4-(3-bromophenyl)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinoline: Into a 500-mL 3-necked round-bottom flask, was placed a solution of 2-((2,4 dichIorobenzyl)(methyl)ammo)-l-(3-bromophenyl)ethanol (25 g, 64 27 πraiol, 1 00 equiv) in dichloromethane (100 mL) This was followed by the addition of sulfuric acid (100 mL) dropwise with stirring at 0-50C The resulting solution was stirred for 4 h at room temperature The resulting solution was diluted with of ice water The pH value of the solution was adjusted to 8 with sodium hydroxide The resulting solution was extracted with 3x300 mL of dichloromethane and the organic layers combined and dried over anhydrous sodium sulfate and concentrated under vacuum The crude product was re-crystallized from petroleum ether ethyl acetate in the ratio of 8 1 This resulted m 15 g (63%) of 4-(3-bromophenyl)-6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoqumoline as a white solid
Figure imgf000148_0001
Intermediate 1.5: 4-(3-(benzylthio)phenyl)-6,8-dichloro-2-methyl- 1 ,2,3,4- tetrahydroisoquinoline: Into a 250-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of potassium carbonate (930 mg, 0 50 equiv) in xylene (50 mL) This was followed by the addition of phenylmethanethiol (2 5 g, 1 50 equiv) dropwise with stirring at O0C The resulting solution was stirred for 1 h at 25°C Into another 100-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was added a solution of 4- (3-bromophenyl)-6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinohne (5 0 g, 1 equiv) in xylene (50 mL), Pd2(dba)3 (300 mg), Xantphos (300 mg) The resulting solution was stirred for 30 mm at 250C and then added to the above reaction solution The mixture was stirred overnight at 14O0C The resulting mixture was concentrated under vacuum The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 100-1 50) This resulted in 2 5 g (45%) of 4-(3 (benzylthio)phenyl) 6,8 dichloro-2- methyl 1,2,3,4-tetrahydroisoqumohne as a yellow oil
Figure imgf000149_0001
Intermediate 1.6: 3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-
5 yl)benzene-l-sulfonyl chloride: Into a 250-mL 3-necked round-bottom flask, was placed a solution of 4-(3-(benzylthio)phenyl)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolme (8 g, 13 53 mmol, 1 00 equiv, 70%) in acetic acid/water (80/8 mL) Cyg) was introduced and the resulting solution was stirred for 1 h at room temperature The resulting mixture was concentrated under vacuum This resulted in 5 0
10 g (90%) of 3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolm-4-yl)benzene-l- sulfonyl chloπde hydrochloride as a yellowish solid
Figure imgf000149_0002
I5 Intermediate 1.7: 2-(2-bromoethyl)isoindo]ine-l,3-dione Into a 500-mL round- bottom flask, was placed a solution of 1 ,2-dibromoethane (30 g, 159 57 mmol, 2 95 equiv) in N.N-dimethylformamide (200 mL) This was followed by the addition of potassium phthalimide (10 g, 54 05 mmol, 1 00 equiv) in several batches The resulting solution was stirred for 24 h at 6O0C The reaction was then quenched by the addition of
20 500 mL of water The resulting solution was extracted with 2x200 mL of ethyl acetate and the organic layers combined and dried over anhydrous sodium sulfate and concentrated under vacuum The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 10) This resulted in 8 g (57%) of 2-(2- bromoethyl)isomdolme-l,3-dione as a white solid
Figure imgf000150_0001
Intermediate 1.8: diethyl 2-(l,3-dioxoisoindolin-2-yl)ethylphosphonate: Into a 50- mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 2-(2-bromoethyl)isoindohne-l,3-dione (8 g, 31 50 πrmol, 1 00 equiv) and tπethyl phosphite (6 2 g, 37 35 mmol, 1 19 equiv) The resulting solution was stirred for 18 h at 13O0C The resulting mixture was concentrated under vacuum The crude product was re-crystallized from ether n-hexane (1 2) This resulted in 5 g (48%) of0 diethyl 2-(l,3-dioxoisomdohn-2-yl)ethylphosphonate as a white solid
Figure imgf000150_0002
Intermediate 1.9: diethyl 2-aminoethylphosphonate: Into a 500-mL round bottom S flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of diethyl 2-(l,3-dioxoisoindohn-2-yl)ethylphosphonate (5 g, 16 08 mmol, 1 00 equiv) in ethanol (200 mL) and hydrazine hydrate (8 g, 160 00 mmol, 9 95 equiv) The resulting solution was stirred for 12 h at room temperature The solids were filtered and the resulting mixture was concentrated under vacuum The residue was applied onto a0 silica gel column and eluted with dichloromethane/methanol (9 1) This resulted in 1 5 g (51%) of diethyl 2-aminoethylphosphonate as colorless oil
Figure imgf000151_0001
Intermediate 1.10: Diethyl 2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsuIfonainido)ethylphosphonate: Into a 50-mL round-bottom flask, was placed a solution of diethyl 2-ammoethylphosphonate (100 mg, 0 55 mmol, 1 00 eqmv) m dichloromethane (10 mL) with TEA (220 rag, 2 18 mmol, 3 94 equiv) This was followed by the addition of 3-(6,8-dichloro-2-methyl- l,2,3,4-tetrahydroisoqumolin-4-yl)benzene-l-sulfonyl chloπde (300 mg, 0 60 mmol, 1 08 equiv, 78%) in several batches The resulting solution was stirred for 2 h at room temperature The reaction progress was monitored by LCMS The resulting mixture was concentrated under vacuum The residue was applied onto a silica gel column with dichloromethane methanol (50 1) This resulted in 0 07 g (24%) of the title compound as a colorless oil
Figure imgf000151_0002
Compound 1 : 2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinoIin-4- yl)phenylsulfonamido)ethylphosphonic acid: To a solution of Intermediate 1 10 (70 mg, 0 13 mmol, 1 00 equiv) in dichloromethane (10 mL) was added bromotnmethylsilane (200 mg, 1 32 mmol, 1004 equiv) The resulting solution was stirred overnight at 4O0C in an oil bath The reaction progress was monitored by LCMS The resulting mixture was concentrated under vacuum To the above was added methanol The resulting mixture was concentrated under vacuum This was followed by the addition of a solution of sodium hydroxide (11 mg, 028 mmol, 2 10 equiv) m methanol (2 mL) The resulting solution was stirred for an additional 1 h at room temperature The resulting mixture was concentrated under vacuum The solid was dned in an oven under reduced pressure This resulted in 52 3 mg (73%) of the title compound as a sodium salt 1H-NMR (300MHz, CD3OD, ppm) 7 82(d, J=I 5Hz, IH), 7 73(s, IH), 7 56(m, IH), 748(d, 7=8 IHz, IH), 7 41 (s, IH), 6 88(s, IH), 4 54(s, IH), 3 97(m, 2H), 3 17(m, 3H), 2 97(m, IH), 2 67(s, 3H), 1 68(m, 2H) MS (ES, m/z) 479 [M+H]+
Example 2
4-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)phenylphosphonic acid
Figure imgf000152_0001
Intermediate 2.1: diethyl 4-nitrophenylphosphonate Into a 100 mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of diethyl phosphonate (3 02 g, 21 88 mmol, 1 10 eqmv) in toluene (10 mL), Pd(PPh3)4 (1 15 g, 1 00 mmol, 0 05 eqmv), TEA (2 21 g, 21 88 mmol, 1 10 equiv), 1 -bromo-4-mtrobenzene (4 g, 19 90 mmol, 1 00 equiv) The resulting solution was stirred for 15 h at 9O0C The solids were filtered out and the resulting mixture was concentrated under vacuum The residue was applied onto a silica gel column and eluted with ethyl acetate/petroleum ether (1 2) This resulted in 3 53 g (68%) of diethyl 4-mtrophenylphosphonate as a yellow liquid
Figure imgf000153_0001
Intermediate 2.2: diethyl 4-aminophenylphosphonate: Into a 50-mL round-bottom flask, was placed a solution of diethyl 4-mtrophenylphosphonate (1 07 g, 4 13 mmol, 1 00 equiv), TEA (3 mL), Palladium carbon (0 025 g) This was followed by the addition of formic acid (2 mL) dropwise with stirring at room temperature The resulting solution was heated to reflux for 3 hr The reaction was then quenched by the addition of 5 mL of water and the solids were filtered out The resulting filtrate was extracted with 5x10 mL of dichloromethane and the organic layers combined and dπed over anhydrous sodium sulfate This resulted in 800 mg (85%) of diethyl 4- ammophenylphosphonate as a white solid
Figure imgf000153_0002
Compound 2: 4-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinoIin-4- yl)phenyl-sulfonamido)phenylphosphonic acid: Compound 2 was prepared m an analogous manner to that of Compound 1 using diethyl 4-aminophenylphosphonate (Intermediate 2 2) as the amine 1H NMR (300MHz, CD3OD, ppm) 7 86(d, IH), 7 69(m, 3H), 7 55(m, 3H), 7 21 (m, 2H), 6 73(s, IH), 4 70(m, 2H), 448(d, IH), 3 79(m, IH), 3 46(m, IH), 3 09(s, 3H) MS (ES, m/z) 527 [M+H]+
Example 3
4-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)benzylphosphonic acid
Figure imgf000154_0001
Intermediate 3.1: diethyl 4-nitrobenzylphosphonate: Into a 250-mL round bottom flask, was placed l-(bromomethyl)-4-mtrobenzene (15 g, 69 77 mmol, 1 OO equiv), tπethyl phosphite (70 mL) The resulting solution was stirred for 2 h at HO0C in an oil bath The resulting mixture was concentrated under vacuum The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 10-1 1) This resulted in 17 g (89%) of the title compound as a yellow oil
Figure imgf000154_0002
Intermediate 3.2: diethyl 4-aminobenzylphosphonate: Into a 100-mL 3-necked round-bottom flask, was placed a solution of diethyl 4-mtrobenzylphosphonate (5 g, 18 32 mmol, 1 00 equiv) in ethanol (50 mL) and a solution of NH4Cl (2 9 g, 54 72 mmol, 2 99 equiv) in water (50 mL) was added This was followed by the addition of Fe (4 1 g, 73 21 mmol, 4 00 equiv), while the temperature was maintained at reflux The resulting solution was heated to reflux for 1 hr The solids were filtered out The resulting mixture was concentrated under vacuum The resulting solution was extracted with 3x20 mL of ethyl acetate and the organic layers combined and dπed over anhydrous sodium sulfate The solids were filtered out The resulting mixture was concentrated under vacuum The residue was applied onto a silica gel column and eluted with ethyl acetate/petroleum ether (1 3) This resulted in 2 5 g (56%) of the title compound as a yellow solid
Figure imgf000155_0001
Compound 3: 4-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)benzylphosphonic acid: Compound 3 was prepared in an analogous manner to that of Compound 1 using diethyl 4-ammobenzylphosphonate (Intermediate 3 2) as the amine 1H-NMR (300MHz, CD3OD, pprri) 7 89(d, J=I 8Hz, IH), 7 61~7 66(m, IH), 7 52-7 54(m, 2H), 7 21-7 20(m, 2H), 7 11(s, IH), 6 95(d, J=S, IHz, 2H), 6 73(s, IH), 4 51-4 59(m, 3H), 3 33(s, IH), 3 03-2 89(m, 6H) MS (ES,
10 m/z) 541 [M+H]+
Example 4
3-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)propylphosphonic acid
I5
Figure imgf000155_0002
Intermediate 4.1: 3-diethyl 3-aminopropylphosphonate: Following the procedures outlined in Example 1, substituting dibromopropane for dibromoethane gave the title 20 compound
Figure imgf000155_0003
Compound 4 3-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)propylphosphonic acid: Compound 4 was prepared in an analogous manner to that of Compound 1 using 3-diethyl 3-aminoρroρylρhosρhonate 5 (Intermediate 4 1) as the amine 1H-NMR (300MHz, CD3OD, ppm) 7 87(d, JS IHz, IH), 7 77(s, IH), 7 61-7 66(m, IH), 7 51-7 54(m, 2H), 6 88(s, IH), 4 77-4 83(m, IH), 4 65(d, /=16 2Hz, IH), 444(d, /-15 6Hz, IH), 3 78-3 84(m, IH), 3 50-3 57(m, IH), 3 08(s, 3H), 2 93-2 97(m, 2H), 1 61-1 72(m, 2H), 1 48-1 59(m, 2H) MS (ES, m/z) 493 [M+H]+ 10
Example 5
(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)methylphosphonic acid
Figure imgf000156_0001
Intermediate 5.1: l,3,5-tribenzyl-l,3,5-triazinane: Into a 100-mL 3-necked round- bottom flask was placed benzylamme (10 g, 93 46 mmol, 1 00 equiv), followed by the addition of formaldehyde (9 0 g, 1 20 equiv, 37%) dropwise with stirring at 0-10cC To 20 the precipitated gum was added 3M aqueous sodium hydroxide (2OmL), and the mixture was stirred After stamdmg in ice for 0 3 h, ether (3OmL) was added, and the mixture stirred until all precipitate dissolved The aqueous phase was separated and extracted with ether The solvents were removed under vacuum to afford 12 g (36%) of 1,3,5 tπbenzyl-l,3,5-tπazinane as colorless oil
25
Figure imgf000156_0002
Intermediate 5.2: diethyl (benzylamino)methylphosphonate: Into a 50-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed l,3,5-tπbenzyl-l,3,5-tπazmane (3 0 g, 8 40 mmol, 1 00 equiv) and diethyl phosphite (3 5 g, 25 36 mmol, 3 00 equiv) The resulting solution was stirred for 3 h at 1000C The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 20 tol 1) This resulted in 2 0 g (90%) of diethyl (benzylammo)methylphosphonate as a colorless oil
CH3COOH
H --OEt EtOH/AcOH O,
Ph. .OEt
H,N_
OEt Pd/C Λ' OEt
Intermediate 5.3: Diethyl aminomethylphosphonate: A 250-mL pressure tank reactor was purged, flushed and maintained with a hydrogen atmosphere, then, was added a solution of diethyl (benzylamino)methylphosphonate (3 5 g, 13 62 mmol, 1 00 equiv) in ethanol (180 mL), acetic acid (10 mL) and Palladium carbon (0 2 g, 0 10 equiv) The resulting solution was stirred for 24 h at 5O0C under 20 atm pressure The solids were filtered out The resulting mixture was concentrated under vacuum This resulted in 2 0 g (crude) of the title compound as brown oil which was used without further purification
Figure imgf000157_0001
Compound 5: (3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)methylphosphonic acid: Compound 5 was prepared in an analogous manner to that of Compound 1 using diethyl aminomethylphosphonate
(Intermediate 5 3) as the amine 1H-NMR (300MHz, CD3OD, ppm) 7 89(d, 7=7 8Hz, IH), 7 74(s, IH), 7 63-7 66(m, IH), 7 57-7 61(m, 2H), 697(s, IH), 4 80-4 89(m, IH), 455-4 67(m, 2H), 3 83-3 89(m, IH), 3 55-3 66(m, IH), 3 02-3 l l(m, 5H) MS (ES, m/z) 465 [M+H]+
Example 6
4-((3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)methyl)benzy]phosphonic acid
OEt P-OEt
H9N O
10
Intermediate 6.1: 4-diethyl 4-(aminomethyl)benzylphosphonate: Following the procedures outlined in Example 1, substituting l,4-bis(bromomethyl)benzene for dibromoethane gave the title compound
Figure imgf000158_0001
Compound 6 4-((3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinoIin-4- yl)phenylsulfonamido)methyl)benzylphosphonic acid: Compound 6 was prepared m an analogous manner to that of Compound 1 using 4-diethyl 4-
20 (ammomethyl)benzylphosphonate (Intermediate 6 1) as the amine 1H-NMR (300MHz, CD3OD, ppm) 7 85~7 88(m, IH), 7 54-7 59(m, 2H), 7 37-7 42(m, 2H), 7 198~7 22(m, 2H), 7 06-7 09(m, IH), 6 77(s, IH), 4 64(m, J=162Hz, IH), 449~4 53(m, IH), 4 37(m, J-16 5, IH), 4 17(s, 2H), 3 45-3 56(m, IH), 3 11-3 27(m, IH), 3 09-3 10(m, 4H), 2 96-2 97(m, IH) MS (ES, m/z) 555 [M+H]+
25
Example 7 3-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolm-4- yl)phenylsulfonamido)propane-l-sulfonic acid
Figure imgf000159_0001
Compound 7: 3-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)propane-l-sulfonic acid: Into a 50-mL round-bottom flask, was placed a solution of 3-aminopropane-l -sulfonic acid (180 mg, 1 29 mmol, 1 00 equiv) in tetrahydrofiiran/water (10/10 niL) with sodium bicarbonate (430 mg, 5 12 mmol) This was followed by the addition of 3-(6, 8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoquinohn-4 yl)benzene-l-sulfonyl chlonde (500 mg, 1 29 mmol, 099 equiv) in several batches The resulting solution was stirred for 4 h at room temperature The reaction progress was monitored by LCMS The pH value of the solution was adjusted to 6 with IM hydrogen chlonde The resulting mixture was concentrated under vacuum The crude product (500 mg) was purified by preparative HPLC to give 26 7 mg of the title compound (4%) as a TFA salt 1H-NMR (300MHz, DMSO, pprri) 10 28(s, IH), 7 53-7 79(m, 6H), 6 83(s, IH), 4 74(s, 2H), 4 51(s, IH), 3 90(s, IH), 3 06 (s, 3H), 2 86-2 93(m, 2H), 2 33-2 44(m, 2H), 1 58-1 63(m, 2H) MS (ES, m/z) 493 [M+H]+
Example 8
2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)-N- (phosphonomethyl)phenylsulfonamido)acetic acid
Intermediate 8.1: ethyl 2-(benzyl((diethoxyphosphoryl)methyl)amino)acetate: Into a 500-mL 3-necked round-bottom flask, was placed a solution of diethyl (benzylamino)methylphosphonate (intermediate 5 2) (12 g, 46 69 mmol, 1 00 equiv) in acetomtnle (150 mL), DIEA (12 g, 2 00 equiv) This was followed by the addition of ethyl 2-bromoacetate (8 4 g, 5030 mmol, 1 10 equiv) dropwise with stirring The mixture was stirred for 30 mm at room temperature The resulting solution was heated to reflux for 6 hr The resulting mixture was cooled to room temperature and concentrated under vacuum The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 20 to 1 5) This resulted in 8 0 g (50%) of ethyl 2- (benzyl((diethoxyphosphoryl)methyl)ammo)acetate as yellow oil
Pd/C V- H /~~& ^" V-COO Et r° COOEt
Intermediate 8.2: ethyl 2-((diethoxyphosphoryl)methylamino)acetate A 250-mL pressure tank reactor was purged, flushed and maintained with a hydrogen atmosphere, then, was added a solution of ethyl 2-
(benzyl((diethoxyphosphoryl)methyl)amino)acetate (8 0 g, 23 32 mmol, 1 00 equiv) in ethanol (180 mL), acetic acid (10 mL), Pd/C (0 9 g) The resulting solution was stirred at 20 atm for 32 h at 5O0C The solids were filtered out, and the resulting mixture was concentrated under vacuum This resulted in 6 0 g (82%) of the acetic acid salt of ethyl
2-((diethoxyphosphoryl)methylamino)acetate as a brown oil
Figure imgf000160_0001
Intermediate 8.3: ethyl 2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin- 4-yl)-N-((diethoxyphosphoryl)methyl)phenyIsulfonamido)acetate: Into a 50-mL round-bottom flask, was placed a solution of ethyl 2- ((diethoxyphosphoryl)methylammo)acetate (320 mg, 1 26 mmol, 1 00 equiv) in pyridine (10 mL) 3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolm-4- yl)benzene-l-sulfonyl chloπde (500 mg, 1 28 mmol, 1 01 equiv) was added and the resulting solution was stirred overnight at room temperature The reaction progress was monitored by LCMS The resulting mixture was concentrated under vacuum The crude product (400 mg) was purified by preparative HPLC to give 200 mg (24%) of the title compound as a TFA salt
Figure imgf000161_0001
Intermediate 8.4 : (3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)-N- (2-ethoxy-2-oxoethyl)phenylsulfonamido)methylphosphonic acid: Into a 50-mL round-bottom flask, was placed a solution of Intermediate 8 3 (200 mg, 0 33 mmol,
1 00 equiv) in dichloromefhane (6 mL) Bromotπmethylsilane (502 mg, 3 30 mmol,
10 01 equiv) was added and the resulting solution was stirred overnight at 4O0C in an oil bath The reaction progress was monitored by LCMS The resulting mixture was concentrated under vacuum The residue was dissolved in 10 mL of methanol The resulting mixture was concentrated under vacuum This resulted m 180 mg (99%) of the title compound as a yellow solid
Figure imgf000162_0001
Compound 8: 2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yI)-N- (phosphonomethyl)phenylsulfonamido)acetic acid: Into a 50-mL round-bottom flask, was placed a solution of (3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin- 4-yl)-N-(2-ethoxy-2-oxoethyl)phenylsulfonamido)methylphosphomc acid
(Intermediate 8 4) (180 mg, O 33 mmol, 1 OO equiv) in tetrahydrofuran/water (5/5 mL) This was followed by the addition of lithium hydroxide (39 mg, 1 62 mmol, 4 97 equiv) in several batches at room temperature The resulting solution was stirred for 4 h at room temperature The reaction progress was monitored by LCMS The resulting mixture was concentrated under vacuum The pH value of the solution was adjusted to 6 with IM hydrogen chloride The resulting mixture was concentrated under vacuum The crude product (150 mg) was purified by preparative HPLC giving 59 2 mg (35%) of the title compound as a TFA salt 1H-NMR (300MHz, DMSO+D2O, ppm) 7 73~7 74(m, IH), 7 67-7 68(m, IH), 7 58-7 62(m, 2H), 749(s, IH), 7 00(s, IH), 4 71~4 75(m, IH), 449(d, J-16 2Hz, IH), 4 33(d, J= 15 9Hz, IH), 4 07(s, 2H), 3 62-3 64(m, IH), 3 45-3 54(m, 2H), 3 31-3 40(m, IH), 2 88(s, 3H) MS (ES, m/z) 523 [MH-H]+
Example 9
2-(3-(6,8-dich]oro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)succimc acid
HCI
NH2 SOCI2 NH2
COOH COOMe
HOOC MeOH MeOOC Intermediate 9.1: Dimethyl 2-aminosuccinate hydrochloride: Into a 100-mL round- bottom flask, was placed a solution of 2-ammosuccinic acid (3 g, 22 56 mmol, 1 00 equiv) in methanol (20 mL) This was followed by the addition of thionyl chloπde (10 g, 8475 mmol, 3 16 equiv) dropwise with stirring at 0-50C The resulting solution was heated to reflux for 2 h in an oil bath The resulting mixture was concentrated under vacuum This resulted in 42 g (95%) of the title compound as a white solid
Figure imgf000163_0001
Intermediate 9.2: Dimethyl 2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)succinate Into a 50-mL round-bottom flask, was placed a solution of dimethyl 2-ammosuccmate hydrochloride (107 mg, 0 54 mmol, 1 00 equiv) in pyridine (5 mL) This was followed by the addition of 3-(6,8- dichloro-2-methyl-l ,2,3,4-tetrahydroisoquinolm-4-yl)benzene-l-sulfonyl chloπde (300 mg, 0 69 mmol, 1 27 equiv, 90%) in several batches The resulting solution was stirred overnight at room temperature The resulting mixture was concentrated under vacuum The residue was applied onto a silica gel column with dichloromethane methanol (50 1) This resulted in 200 mg (72%) of the title compound as a colorless oil
Figure imgf000163_0002
Compound 9: 2-(3-(6,8-dichloro-2-methy]-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)succinic acid: Into a 50-mL round-bottom flask, was placed a solution of Intermediate 9 2 (100 mg, 0 19 mmol, 1 00 equiv) in tetrahydrofuran (5 mL) and water (5 mL) This was followed by the addition of LiOH (23 mg, 096 mmol, 493 equiv) in several batches at room temperature The resulting solution was stirred for 2 h at room temperature The reaction progress was monitored by LCMS The resulting mixture was concentrated under vacuum The pH value of the solution was adjusted to
6 with hydrogen chloπde (1 mol/L) The solids were collected by filtration The crude product (200 mg) was purified by preparative HPLC to give 12 1 mg (10%) the title compound as a TFA salt 1H-NMR (300MHz, CD3OD, ppm) 7 89(d, J=I 2Hz, IH),
7 80(d, J-6 3Hz, IH), 7 64-7 52(m, 3H), 6 95(s, IH), 4 78-4 70(m, 2H), 4 55-4 50(m, IH), 4 23~4 17(m, IH), 3 87-3 82(m, IH), 3 63-3 57(m, IH), 3 12(s, 3H), 2 79-2 65(m, 2H) MS (ES, tn/z) 487 [M-CF3COOH+H]+
Example 10
2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethylphosphonic acid
Figure imgf000164_0001
Intermediate 10.1: 2-bromo-l-(4-bromophenyl)ethanone: Into a 250-mL 3-necked round-bottom flask, was placed a solution of 1 (4-bromophenyl)ethanone (10 0 g, 50 25 mmol, 1 00 equiv) in acetic acid (50 mL) This was followed by the addition of a solution of bromine (8 2 g, 1 05 equiv) in acetic acid (50 mL) dropwise with stirring at 6O0C over 90 min The resulting solution was stirred for 3 h at 6O0C The resulting mixture was concentrated under vacuum The crude product was re-crystallized from petroleum ether/ethyl acetate m the ratio of 7 1 This resulted in 9 3 g (67%) of the title compound as a yellow solid
Figure imgf000165_0001
Intermediate 10.2: l-(4-bromophenyl)-2-((2,4- dichlorobenzyl)(methyl)amino)ethanone: Into a 250-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 2-bromo-l-(4-bromophenyl)ethanone (9 3 g, 33 45 mmol, 1 00 equiv) in dioxane (100 mL), tπethylamine (5 0 g, 1 50 equiv), and (2,4-dichlorophenyl)-N- methylmethanamine (64 g, 33 68 mmol, 1 00 equiv) The resulting solution was stirred for 2 h at 250C The solids were filtered out The filtrate was used for next step directly
Figure imgf000165_0002
Intermediate 10.3: 2-((2,4-dichIorobenzyl)(methyl)amino)-l-(4- bromophenyl)ethanol: Into a 500-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of the crude Intermediate 10 2 m fresh methanol (100 mL) This was followed by the addition of sodium borohydnde (2 5 g, 65 79 mmol, 2 00 equiv) in several batches at 0-5cC The resulting solution was stirred for 1 h at 250C The reaction was then quenched by the addition of sat NH4CI The resulting mixture was concentrated under vacuum The resulting solution was extracted with EtOAc (2x100 mL) and the organic layers combined and concentrated under vacuum The crude product was re-crystalhzed from petroleum ether/ethyl acetate(60 mL) in the ratio of 7 1 This resulted in 6 5 g (50%) of the title compound as a white solid MS (ES, m/z) 390 [M+H]+
Figure imgf000166_0001
Intermediate 10.4: 4-(4-bromophenyl)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinoline Into a 50-mL 3-necked round-bottom flask, was placed a solution of 2-((2,4-dichlorobenzyl)(methyl)ammo)-l-(4-bromophenyl)ethanol (1 0 g, 2 57 mmol, 1 00 eqmv) m dichloromethane (3 mL) This was followed by the addition of cone H2SO4 (2 mL) dropwise with stirring at 0-50C The resulting solution was stirred for 3 h at 200C The reaction was then quenched by the addition of water/ice The pH value of the solution was adjusted to 9 with sodium hydroxide The resulting solution was extracted with dichloromethane (2x30mL) and the organic layers combined and dπed over anhydrous sodium sulfate and concentrated under vacuum This resulted in 09 g of the title compound which was used without further purification MS (ES, m/z) 372 [M+H]+
Figure imgf000166_0002
Intermediate 10.5: 4-(4-(benzyIthio)phenyl)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinoline: Into a 250-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed K2CO3 (800 mg, 0 50 equiv) and xylene (50 mL) This was followed by the addition of phenylmethanethiol (1 75 g, 1 00 equiv) dropwise with stirring at O0C The resulting mixture was then allowed to warm to room temperature and stirred for 1 h Into another 250-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 4-(4-bromoρhenyl)-6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinoline (4 8 g, 0 80 equiv), Xantphos (200 mg, 0 08 equiv) and Pd2(dba)3 (200 mg,0 08 equiv) in xylene (30 mL) The mixture was stirred at room temperature for 20 mm and transferred to the previously formed potassium thiolate The dark solution was then purged with nitrogen and heated to 13O0C for 15 h After cooling to room temperature, the mixture was concentrated under reduced pressure The crude product was then purified by silica gel chromatography with ethyl acetate/petroleum ether (1 80~l 50) to afford 1 8 g (30%) of the title compound as yellow oil MS (ES, mJz) 414 [M+H]+
Figure imgf000167_0001
Compound 10.6: 4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)benzene-l-sulfonyl chloride: Into a 50-mL 3-necked round-bottom flask, was placed a solution of 4-(4-(benzylfhio)phenyl)-6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoqumolme (250 mg, 0 60 mmol, 1 00 equiv) m acetic acid (8 mL), water (1 mL) To the above Q2(g) was introduced and the resulting solution was stirred for 30 mm at 250C The resulting mixture was concentrated under vacuum This resulted in 200 mg (85%) of the title compound as a yellow solid MS (ES, m/z) 390 [M- HCHH]+
Figure imgf000167_0002
Compound 10: 2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethylphosphonic acid: Following the procedures outlined in Example 1 , 4-(6,8-dichloro-2-methyl- 1 ,2,3 ,4-tetrahydroisoqumolm-4-yl)benzene- 1 - 5 sulfonyl chloπde (intermediate 10 6) was converted to compound 10 Purification by preparative HPLC gave a TFA salt of the title compound as a white solid 1H-NMR (CD3OD, 300MHz, ppm) 7 93(d, J=S, 4Hz, 2H), 7 58~7 51(m, 3H), 6 89(s, IH), 4 89~4 80(m, 2H), 4 56~4 51(m, IH), 3 95-3 90(m, IH), 3 69-3 65(m, IH), 3 21-3 10(m, 5H), 2 01-1 89(m, 2H) MS (ES, m/z) 479 [M+H]+ 10
Example 11
(4-(6,8-dich]oro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)methylphosphonic acid
Figure imgf000168_0001
Compound 11 : (4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)methylphosphonic acid: Following the procedures outlined in Example 1 , compound 11 was made using 4-(6,8-dichloro-2-methyl-l, 2,3,4-
20 tetrahydroisoqumolin-4-yl)benzene-l -sulfonyl chloπde (intermediate 10 6) and diethyl ammomethylphosphonate (intermediate 5 3) Purification by preparative HPLC gave a TFA salt of the title compound 1H NMR (300MHz, DMSOD2O, ppm) 7 87(d, J=8 4Hz, 2H),7 68(d, J=I 5Hz, IH), 7 48(d, J=9 4Hz, 2H), 6 80(s, IH), 4 74-4 66(m, IH), 4 46~4 40(m, IH), 3 82-3 77(m, IH), 3 69-3 39(m, IH), 3 01(s, 3H),
25 2 91-2 74(m, 2H) MS 465 [M+H]+ Example 12
3-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)propylphosphonic acid
Figure imgf000169_0001
Compound 12: 3-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)propylphosphonic acid: Following the procedures outlined in
10 Example 1, compound 12 was made using 4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzene-l-sulfonyl chloride (intermediate 10 6) and 3- diethyl 3-ammopropylphosphonate (intermediate 4 1) Purification by preparative HPLC gave a TFA salt of the title compound 1H-NMR (300MHz, CD3OD, ppm) 7 90(d, J-8 4, 2H), 7 55(s, IH), 7 46(d, J=& IHz, 2H), 6 88(s, IH), 4 77-4 82(m, IH),
15 4 71(d, J 16 2Hz, IH), 4 47(d, J= 15 9Hz, IH), 3 80-3 86(m, IH), 3 54-3 61 (m, IH), 3 l l(s, 3H), 2 95-2 99(m, 2H), 1 53-1 71(m, 4H) MS 493 [M+H]+
Example 13
(4-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- 20 yl)phenyl$ulfonamido)phenyl)methylphosphonic acid
Figure imgf000170_0001
Compound 13: (4-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)phenyl)methylphosphonic acid: Following the procedures outlined in Example 1, compound 13 was made using 4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoqumolin-4-yl)benzene 1-sulfonyl chloride (intermediate 10 6) and 4- ammobenzylphosphonate (intermediate 3 2) Puπfication by preparative HPLC gave a TFA salt of the title compound 1H-NMR (300MHz, DMSO+D2O, ppm) 7 69(d, J-S 4Hz, 2H), 746-746(m, IH), 7 34(d, 7-8 4Hz, 2H), 7 07(d, J=I 8Hz, 2H), 6 94(d, J=S IHz, 2H), 6 71-6 71(m, IH), 4 36-440(m, IH), 3 65-3 80(m, 2H), 2 95-3 01(m, IH), 2 72-2 79(m, 3H), 2 41 (s, 3H) MS (ES, m/z) 541 [M+H]+
Example 14
(4-((4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)methyl)phenyl) methylphosphonic acid
Figure imgf000170_0002
Compound 14: (4-((4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)methyl)phenyl) methylphosphonic acid: Following the procedures outlined in Example 1, compound 14 was made using 4-(6,8-dichloro-2- methyl- 1 ,2,3 ,4-tetrahydroisoquinolm-4-yl)benzene- 1 -sulfonyl chlonde (intermediate 10 6) and 4-(aminomethyl)benzylρhosphonate (intermediate 6 1) Purification by preparative HPLC gave a TFA salt of the title compound 1H-NMR (300MHz, DMSO+D2O, ppm) l l\{ά, J 8 4Hz, 2H), 7 50(m, IH), 7 40(d, J 8 4Hz, 2H), 7 06~7 15(m, 4H), 6 86-6 87(m, IH), 438~440(m, IH), 3 95(s, 2H), 3 75(d, J-16 2Hz, IH), 3 53(m, IH), 2 85-2 92(m, 3H), 2 69-2 75(m, IH), 241 (s, 3H) MS (ES, m/z) 555 [M+H]+
Example 15
3,3'-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinoIin-4- yl)phenylsulfonylazanediyl)dipropanoic acid
Figure imgf000171_0001
Intermediate 15.1: 2-((2,4-dichlorobenzyl)(methyl)amino)-l-phenylethanone: Into a 50-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 2-bromo 1 -phenyl ethanone (1 g, 5 05 mmol, 1 00 equiv) in 1,4-dioxane (20 mL) and (2,4-dichlorophenyl)-N-methylmethanamme (1 1 g, 5 82 mmol, 1 15 equiv) Tπethylamine (2 g, 19 80 mmol, 3 92 equiv) was added dropwise with stirring at 2O0C The resulting solution was stirred for 1 h at 2O0C in an oil bath The solids were filtered out The resulting mixture was concentrated under vacuum The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 50) This resulted in 1 4 g (90%) of the title compound as a yellow oil
Figure imgf000172_0001
Intermediate 15.2: 2-((2,4-dichlorobenzyI)(methyl)atnino)-l-phenylethanol: Into a 250 ml 3-necked roundbottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 2-((2,4-dichlorobenzyl)(methyl)ammo)-l- phenylethanone (4 3 g, 14 01 mmol, 1 00 equiv) in methanol (50 mL) This was followed by the addition of NaBH4 (1 5 g, 3947 mmol, 2 82 equiv) m several batches at O0C The resulting solution was stirred for 30 mm at 00C in a water/ice bath The reaction was then quenched by the addition of 20 mL of acetone The resulting mixture
10 was concentrated under vacuum The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 80-1 20) This resulted in 3 4 g (79%) of the title compound as a white solid
Figure imgf000172_0002
I5
Intermediate 15.3: 6,8-dichloro-2-methyI-4-phenyl-l,2,3,4-tetrahydroisoquinoline:
Into a 100-mL 3-necked round-bottom flask, was placed a solution of 2-((2,4- dichlorobenzyl)(methyl)ammo)-l -phenyl ethanol (3 4 g, 11 00 mmol, 1 00 equiv) m dichloromethane (15 mL) This was followed by the addition of sulfuric acid (15 mL)
20 dropwise with stirring at 00C The resulting solution was stirred for 2 h at O0C in a water/ice bath The pH value of the solution was adjusted to 7 with IM sodium hydroxide The resulting solution was extracted with ethyl acetate (3x60mL) and the combined organic layers dπed over anhydrous sodium sulfate and concentrated under vacuum The residue was applied onto a silica gel column with petroleum ether ethyl
25 acetate (80 1) This resulted in 1 6 g (50%) of the title compound as a colorless oil
Figure imgf000173_0001
Intermediate 15.4: 4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)benzenesulfonamide: Into a 100-mL 3-necked round-bottom flask purged and 5 maintained with an inert atmosphere of nitrogen, was placed chlorosulfonic acid (4 mL) This was followed by the dropwise addition of a solution of 6,8-dichloro-2- methyl-4-phenyl-l,2,3,4-tetrahydroisoqmnolme (1 6 g, 5 5 mmol, 1 00 equiv) in dichloromethane (30 mL) at O0C The resulting solution was stirred for 1 h at O0C in a water/ice bath and for an additional 1 h at 250C in an oil bath To this was added
10 chlorosulfonic acid (16 mL) dropwise at 250C The resulting solution was stirred for an additional 1 h at 250C To the resulting mixture was cooled to 0°C and aqueous ammonia (120 mL) was added dropwise The resulting solution was stirred for an additional 3 h 9O0C in an oil bath The resulting mixture was concentrated under vacuum The residue was dissolved in 20 mL of water The resulting solution was
15 extracted with dichloromethane (3x30mL) and the combined organic layers concentrated under vacuum The residue was applied onto a silica gel column with dichloromethane/methanol (100 1) The crude product (0 5 g) was purified by preparative HPLC to give 53 mg (3%) of the title compound as a TFA salt 1H-NMR (300MHz,CDCl3, ppm) 7 89(1H, d,J-8 4Hz), 7 35(2H, d,J-8 4Hz), 7 30(1H, m),
20 6 77(1H, s), 4 87(1H, s), 4 39(1H, s), 3 69(2H, m), 2 98(1 H, t), 2 67(1 H, dd), 2 55(3H, s) MS (ES, m/z) 371 [M+H]+
Figure imgf000174_0001
Intermediate 15.5: dimethyl 3,3'-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonyIazanediyl)dipropanoate: Into a 50-mL 3- necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumohn-4- yl)benzenesulfonamide (Compound 15 4, 100 mg, 027 mmol, 1 00 equiv) in acetomtnle (5 mL) Methyl but-3-enoate (40 mg, 040 mmol, 1 48 equiv) was added, along with 1,8-Diazabicyclo[5 4 0]undec-7-ene (DBU, 20 mg, 0 13 mmol, 049 equiv) The resulting solution was stirred overnight at 25°C m an oil bath Removing the solvent under vacuum gave the title compound which was used without further pun fi cation
Figure imgf000174_0002
Compound 15: 3,3'-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonylazanediyl)dipropanoic acid: Into a 50-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of Intermediate 15 5 (140 mg, 026 mmol, 1 00 equiv, theoretical yield) in tetrahydrofuran(5 mL) and water (5 mL) LiOH (20 mg, 0 83 mmol, 3 23 equiv) was added and the resulting solution was stirred for 1 h at room temperature The resulting mixture was concentrated under vacuum The residue was applied onto a silica gel column with dichloromethane/methanol (100 1-20 1) This resulted m 0 015 g (11%) of the title compound as a white solid 1H-NMR (300MHz, CD3OD, ppm). 7 84(d, 7=8 IHz, 2H), 7 41 (d, J=8 4Hz, 2H), 7 35(s, IH), 6 84(s, IH), 4 39(t, IH), 3 77(d, IH), 3 67(d, IH), 3 45(m, IH), 3 33(m, 4H), 2 69(d, IH), 3 0(m, IH), 2 47(m, 6H) MS (ES, m/z) 515 [M+H]+
Example 16 N,N',N"-(2,2',2"-nitrilotris(ethane-2,l-diyl))tris(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide)
Figure imgf000175_0001
Compound 16: N,N',N"-(2,2',2"-nitrilotris(ethane-2,l-diyl))tris(3-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide): To a solution of 3- (6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinohn-4-yl)benzene-l-sulfonyl chloπde (intermediate 1 6) (lOOmg, 0235mmol) in DMF (1 5mL) was added TEA (94 94mg, 0 94mmol) and a solution of Nl,Nl-bis(2-aminoethyl)ethane-l,2-diaimne (11 45mg, 0 0783mmol) in 0 ImL DMF The reaction was stirred for 40 minutes at which point LCMS indicated no starting matenal remained The solvent was removed and the residue dissolved in 50% acetic acid in water and purified by preparative HPLC to yield the title compound (25 4mg) as a TFA salt 1H-NMR (400MHz, d6-DMSO) 57 77 (s, IH), 7 75 (s, IH), 7 64 (s, IH), 7 59 (m, 3H), 6 76 (s, IH), 470 (m, IH), 4 38 (m, IH), 3 90 (brm, 8H), 3 26 (m, IH), 3 95 (s, 3H), 2 65 (m, 2H) MS (m/z) 1210 01 (M+H)
Example 17
N,N'-(2,2'-(ethane-l,2-diylbis(oxy))bis(ethane-2,l-diyl))bis(3-(6,8-dichIoro-2- methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesuIfonamide)
Figure imgf000176_0001
Compound 17: N,N'-(2,2'-(ethane-l,2-diylbis(oxy))bis(ethane-2,l-diyl))bis(3-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide): To a solution of 2,2'-(ethane-l,2-diylbis(oxy))diethanamme (26 17mg, 0 176mmol) in chloroform (0 223mL) at 00C was added dnsopropylethylamine (DIEA, 182mg, 1 412mmol) and a solution of 3-(6,8-dichloro 2 methyl- l,2,3,4-tetrahydroisoqumolm-4- yl)benzene-l-sulfonyl chloπde (intermediate 1 6) (150mg, 0 353mmol) in chloroform (0 706mL) The resulting solution was stirred for 10 minutes at which point the solvent was removed and the residue taken up m 50% isopropanol/water mixture and purified by preparative HPLC The title compound was obtained (44 5mg) as a TFA salt 1H- NMR (400MHz, CD3OD) 57 87 (d, IH), 778 (d, IH), 7 64 (t, IH), 7 55 (d, IH), 7 51 (d, IH), 6 81 (s, IH), 4 47 (d, IH), 3 83 (dd, IH), 3 59 (t, IH), 3 43 (m, 2H), 3 12 (s, 4H), 3 01 (q, 2H) MS (m/z) 857 17 (M+H)
Example 18
N,N'-(l,4-phenylenebis(methylene))bis(3-(6,8-dichIoro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonainide)
Figure imgf000177_0001
Compound 18: N,N'-(l,4-phenylenebis(methylene))bis(3-(6,8-dichloro-2-methyl- l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide): Following the procedures outlined in Example 17, compound 18 was made using 1,4-phenylenedimethanaτnme as the amine Purification by preparative HPLC gave the title compound as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 87 (d, 2H), 7 61 (s, 2H), 7 52 (m, 4H), 7 49 (d, 2H), 7 09 (s, 4H), 6 82 (s, 2H), 4 78 (m, 7H), 443 (d, 2H), 4 00 (s, 4H), 3 82 (dd, 2H), 3 51 (t, 2H), 3 11 (s, 6H) MS (m/z) 845 03 (M+H)
Example 19
N,N'-(butane-l,4-diyl)bis(3-(6,8-dichloro-2-methy]-l,2,3,4-tetrahydroisoquinoIin-4- yl)benzenesulfonamide)
Figure imgf000177_0002
Compound 19: N,N'-(butane-l,4-diyl)bis(3-(6,8-dichIoro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide): Following the procedures outlined in Example 17, compound 19 was made using butane- 1,4-diamine as the amine Purification by preparative HPLC gave the title compound as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 85 (d, 2H), 7 80 (s, 2H), 7 63 (t, 2H), 7 54 (t, 4H), 6 82 (s, 2H), 449 (d, IH), 3 88 (dd, 2H), 3 58 (t, 2H), 3 14 (s, 6H), 2 81 (m, 4H), 1 42 (m, 4H) MS (m/z) 797 19 (M+H) Example 20
N,N'-(dodecane-l,12-diyl)bis(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide)
Figure imgf000178_0001
Compound 20: N,N'-(dodecane-l,12-diyl)bis(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide): Following the procedures outlined in Example 17, compound 20 was made using dodecane-l ,12-diamine as the amine Purification by preparative HPLC gave the title compound as a TFA salt 1H-NMR (400MHz, CD3OD) 57 85 (d, 2H), 7 71 (s, 2H), 7 63 (t, 2H), 7 54 (m, 4H), 6 81 (s, 2H), 4 74 (m, 2H), 4 51 (d, 2H), 3 86 (dd, 2H), 3 29 (t, 2H), 3 13 (s, 7H), 2 79 (t, 4H), 1 39 (m, 4H), 1 22 (m, 20H) MS (Wz) 909 28 (M+H)
Example 21
N,N',N",N'"-(3,3',3",3'"-(butane-l,4-diylbis(azanetriyl))tetrakis(propane-3,l- diyl))tetrakis(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)benzenesulfonamide)
Figure imgf000179_0001
Compound 21: N,N',N",N"'-(3,3',3",3'"-(butane-l,4- diylbis(azanetriyl))tetrakis(propane-3,l-diyl))tetrakis(3-(6,8-dichloro-2-methyl- l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide): To a solution of 3-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolm-4-yl)benzene-l-sulfonyl chlonde
(intermediate 1 6) (150mg, 0 352mmol) m THF/H2O (0 704mL, 50% v/v) was added DIEA (181 6mg, 1 41mmol) and finally Nl,Nl'-(butane-l,4-diyl)bis(Nl-(3- ammopropyl)proρane-l,3-diamine) (27 94mg, 0 08825mmol) The reaction mixture was stirred vigorously for 1 hour at which point the solvent was removed The resulting residue was brought up in 50% acetomtπle/water and purified by preparative HPLC to give the title compound (117mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ7 85 (d, 2H), 7 78 (s, 2H), 7 62 (t, 2H), 7 36 (m, 4H), 6 79 (s, 2H), 4 78 (m, 4H), 4 47 (d, 2H), 3 86 (dd, 2H), 3 55 (t, 2H), 3 12 (s, 6H), 2 94 (m, 4H), 1 90 (m, 4H), 1 85 (m, 2H) MS (m/z) 1732 90 (M+H)
Example 22
N,N'-(butane-l,4-diyl)bis(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)benzenesulfonamide)
Figure imgf000180_0001
Compound 22: N,N'-(butane-l,4-diyl)bis(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide): To a solution of 4-(6,8-dichloro-2- methyl- 1 ,2,3,4-tetrahydroisoqumolin-4-yl)benzene- 1 -sulfonyl chlonde (intermediate 10 6) (150mg, 0 353mmol) in chloroform (0706mL) was added DIEA (182mg, 1 412mmol) and a solution of butane- 1,4-diamme (15 5mg, 0 176mmol) m chloroform (0 176mL) The reaction was stirred overnight at which point the solvent was removed and the resulting residue brought up in 50% IPA/H2O Purification by preparative HPLC gave the title compound (18 4mg) as a TFA salt 1H-NMR (400MHz, CD3OD) 57 86 (d, 4H), 7 53 (s, 2H), 7 45 (d, 4H), 6 84 (s, 2H), 4 73 (m, 3H), 446(d, 2H), 3 86 (dd, 2H), 3 57 (t, 2H), 3 12 (s, 6H), 2 84 (m, 4H), 1 41 (m, 4H) MS (m/z) 797 15 (M+H)
Example 23
N,N'-(dodecane-l,12-diyl)bis(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide)
Figure imgf000180_0002
Compound 23: N,N'-(dodecane-l,12-diyl)bis(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide): Following the procedures outlined in Example 22, compound 23 was made using dodecane-l,12-diamine as the amine Purification by preparative HPLC gave the title compound as a TFA salt 1H-NMR (400MHz, CD3OD) 7 89 (d, 4H), 7 54 (m, 2H), 742 (m, 4H), 6 82 (s, 2H), 4 85 (m, 3H), 4 72 (d, 2H), 3 85 (dd, 2H), 3 59 (t, 2H), 3 13 (m, 8H), 2 85 (m, 4H), 1 89 (m, 5H), 1 33 (m, 23H) MS {m/z) 90921 (M+H)
Example 24
N,N',N"-(2,2',2"-nitraotris(ethane-2,l-diyl))tris(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide)
Figure imgf000181_0001
Compound 24: N,N',N"-(2,2',2"-nitrilotris(ethane-2,l-diyl))tris(4-(6,8-dichloro-2- methyI-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide): To a solution of 4- (6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinohn-4-yl)benzene-l sulfonyl chlonde (intermediate 10 6) (150mg, 0 353mmol) in THF/H2O solution (50% v/v, 0 704mL) was added DIEA (182 2mg, 1 412mmol) and Nl,Nl-bis(2-aminoethyl)ethane-l,2- diamine (17 0mg, O l lδmmol) The reaction was stirred vigorously at room temperature for 40 minutes at which point the solvent was removed The resulting residue was dissolved in acetomtnle/water (50% v/v) and purified by preparative HPLC to give the title compound (57 6mg) as a TFA salt 1H-NMR (400MHz, CD3OD) 7 94 (d, 6H), 7 51 (t, 9H), 6 83 (s, 3H), 4 78 (m, 6H), 4 45( d, 3H), 3 83 (dd, 3H), 3 49 (t, 3H), 3 30 (m, 6H), 3 29 (m, 21H), 3 12 (s, 9H) MS (m/z) 1208 09 (M+H)
Example 25
N,N1,N",N'"-(3,3l,3",3'"-(butane-l,4-diylbis(azanetriyl))tetrakis(propane-3,l- diyl))tetrakis(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)benzenesulfonamide)
Figure imgf000182_0001
Compound 25: N,N',N",N'"-(3,3',3",3'"-(butane-l,4- diylbis(azanetriyl))tetrakis(propane-3,l-diyl))tetrakis(4-(6,8-dichloro-2-methyl- l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide): Following the procedure outlined in Example 24, Compound 25 was made using Nl,Nl'-(butane-l,4- diyl)bis(Nl-(3-armnoproρyl)propane-l,3-diarmne) as the amine Purification by preparative HPLC gave the title compound as a TFA salt 1H-NMR (400MHz, CD3OD) 7 88 (d, 8H), 7 51 (s, 4H), 7 48 (d, 8H), 6 81 (s, 4H), 4 75 (m, 8H), 4 47 (d, 4H), 3 85 (dd, 4H), 3 58 (t, 4H), 3 13 (s, 12H), 2 98 (t, 8H), 1 97 (m, 8H), 1 88 (m, 4H) MS (Wz) 1733 02 (M+H)
Example 26 N,N'-(l,4-phenylenebis(methylene))bis(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoqulnolin-4-yl)benzenesulfonamide)
Figure imgf000183_0001
Compound 26: N,N'-(l,4-phenylenebis(methylene))bis(4-(6,8-dichloro-2-methyl- l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide): Following the procedure outlined in Example 24, compound 26 was made using 1,4-phenylenedimethanamine as the amine Purification by preparative HPLC gave the title compound as a TFA salt 1H-NMR (400MHz, CD3OD) 7 76 (d, 4H), 7 54 (s, 2H), 7 39 (d, 4H), 7 08 (s, 4H), 6 82 (s, 2H), 4 72 (m, 3H), 447 (d, 2H), 4 07 (s, 4H), 3 88 (dd, 2H), 3 61 (t, 2H), 3 16 (s, 6H) MS (m/z) 845 07 (M+H)
Example 27
N,N'-(2,2'-(ethane-l,2-diylbis(oxy))bis(ethane-2,l-diyl))bis(4-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide)
Figure imgf000183_0002
Compound 27: N,N'-(2,2'-(ethane-l,2-diylbis(oxy))bis(ethane-2,l-diyl))bis(4-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesu]fonamide):
Following the procedure outlined in Example 24, compound 27 was made using 2,2'- (ethane-l,2-diylbis(oxy))diethanamine as the amine Purification by preparative HPLC gave the title compound as a TFA salt 1H-NMR (400MHz, CD3OD) 7 89 (d 4H), 7 52 (s, 2H), 7 47 (d, 4H), 6 82 (s, 2H), 4 77 (m, 4H), 4 47 (d, 2H), 3 86 (dd, 2H), 3 59 (t, 2H), 3 43 (t, 8H), 3 13 (s, 6H), 3 06 (t, 4H) MS (m/z) 857 15 (M+H) Example 28
N-(2-(2-(2-(2-amlnoethoxy)ethoxy)ethoxy)ethyI)-3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesuIfonamide
Figure imgf000184_0001
Intermediate 28.1 N-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro- 2-methyI-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide: To a solution of 3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumohn-4-yl)benzene-l-sulfonyl chloπde (intermediate 1 6) (600mg, 1.41mmol) in chloroform (2 82mL) was added DIEA (545 7mg, 4 24mmol) and 2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethanarmne (616 3mg, 2 82mmol) The reaction was stirred overnight at which point the mixture was diluted with 5OmL DCM and washed with NaHCO3 (5OmL) The aqueous layer was extracted with DCM (2x50mL) and the combined organic fractions washed with water (20OmL), bnne (20OmL), and dπed over Na2SU4 Removing the solvent gave the title compound as an oil which was used without further purification
Figure imgf000184_0002
Compound 28: N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoquinolin-4-y])benzenesulfonamide: N-(2-(2-(2-(2- azidoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoqumohn-4-yl)benzenesulfonamide (intermediate 28 1) (1 035g, assume 1 41mmol) was dissolved in a 10 1 THF water solution (26 5mL) and placed under N2 PMβ3 (165mg, 2 18mmol) was added and the reaction stirred overnight The solvent was removed and the resulting residue brought up in EtOAc (10OmL) and washed with NaHCθ3 (10OmL) and bπne (10OmL) After drying the organic layer over Na2SCv, the solvent was removed to give 446mg of the title compound (58% over two steps) as an oil A portion of the crude product was purified by preparative HPLC to give the title compound as a TFA salt 1H-NMR (400 mHz, CD3OD) δ 7 87 (m, IH), 7 73 (m, IH), 7 67 (t, j-7 7 Hz, IH), 7 54 (m, 2H), 6 82 (s, IH), 4 8-46 (m, 4H), 4 46 (m, IH), 3 86 (m, IH), 3 69 (m, 2H), 3 66 (s, 3H), 3 61 (m, 2H), 3 55 (m, 2H), 3 12 (m, 4H), 3 03 (t, j-5 4 Hz, IH) MS (m/z) 546 18 (M+H)
Example 29
Nl,N8-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)octanediamide
Figure imgf000185_0001
Compound 29: Nl,N8-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)octanediamide: To a solution of N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl) 3-(6,8-dichloro-2-methyl-l,2,3,4 tetrahydroisoqumolin-4 yl)benzenesulfonamide (compound 28) (54 5mg, 0 lmmol) in DMF (0 2OmL) was added DIEA (15 5mg, 0 12mmol) and bis(2,5 dioxopyrrolidm-1- yl) octanedioate (18 4mg, 0 05mmol) The reaction was stirred at room temperature for 3 hours at which point an additional 0 03mmol of compound 28 was added After a further hour the solvent was removed and the resulting residue dissolved in acetomtπle/water (1 1) and puπfied by preparative HPLC to give the title compound (17 4mg) as a TFA salt 1H-NMR (400MHz, CD3OD) 7 89 (d, 2H), 7 78 (s, 2H), 7 64 (t, 2H), 7 52 (m, 4H), 6 83 (s, 2H), 4 81 (m, 4H), 4 45 (d, 2H), 3 89 (dd, 2H), 3 61 (m, 18H), 3 55 (m, 10H), 3 47 (m, 5H), 3 33 (m, 5H), 3 14 (s, 7H), 3 04 (t, 4H), 2 16 (t, 4H), 1 55 (m, 4H), 1 29 (m, 4H) MS (m/z) 1231 87 (M+H)
Example 30
2-(N-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquiπolin-4- yl)phenyl)sulfamoylamino)ethylphosphonic acid
Figure imgf000186_0001
Intermediate 30.1: l-(4-aminophenyl)ethanone Into a 100-mL 3-necked round- bottom flask, was placed a solution of l-(4-mtrophenyl)ethanone (6 g, 36 36 mmol,
1 00 equiv) in ethanol(100 mL), water(15 mL) This was followed by the addition of
NH4Cl (3 85 g, 72 64 mmol, 2 00 equiv) in several batches To this was added Fe
(10 18 g, 181 79 mmol, 5 00 equiv) m several batches, while the temperature was maintained at reflux The resulting mixture was heated to reflux for 2 h The solids were filtered out and the resulting filtrate was concentrated under vacuum The residue was diluted with 50 mL of water The resulting solution was extracted with 3x50 mL of ethyl acetate and the organic layers combined and dπed over anhydrous sodium sulfate and concentrated under vacuumto give 3 1 g (60%) of 1 (4-aminophenyl)ethanone as a yellow solid
Figure imgf000187_0001
Intermediate 30.2: N-(4-acetylphenyI)acetamide: Into a 100-mL 3-necked round- bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of l-(4-aminophenyl)ethanone (3 1 g, 22 96 mmol, 1 00 equiv) in dichloromethane (30 mL), tnethylamme (4 64 g, 45 94 mmol, 2 00 equiv) This was followed by the addition of acetyl chloπde (1 79 g, 22 95 mmol, 1 00 equiv) dropwise with stirring at O0C The resulting solution was stirred for 30 mm at O0C The reaction was then quenched by the addition of 2 mL of water The resulting mixture was washed with 3x50 mL of saturated aqueous sodium chlonde The mixture was dπed over anhydrous sodium sulfate and concentrated under vacuum to give 3 O g (74%) of N-(4- acetylρhenyl)acetamide as a white solid
Intermediate 30.3: N-(4-(2-bromoacetyl)phenyl)acetamide: Into a 100-mL 3- necked round-bottom flask, was placed a solution of N-(4-acetylphenyl)acetamide (1 g, 5 65 mmol, 1 00 equiv) in acetic acid (10 mL) This was followed by the addition of a solution of bromine (910 mg, 5 69 mmol, 1 01 equiv) in acetic acid (2 mL) dropwise with stirring at 5O0C The resulting solution was stirred for 1 5 h at 5O0C The reaction was then quenched by the addition of 100 mL of water/ice The solids were collected by filtration and dπed under vacuum This resulted m 0 5 g (33%) of N-(4-(2- bromoacety])phenyl)acetamide as a white solid
Figure imgf000188_0001
Intermediate 30.4: N-(4-(2-((2,4- dichlorobenzy])(methyl)amino)acetyl)phenyl)acetamide: Into a 100-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of N-(4-(2-bromoacetyl)phenyl)acetamide (1 g, 3 91 mmol, 1 00 equiv) in 1,4-dioxane (40 mL) This was followed by the addition of tnethylamine (1 58 g, 15 64 mmol, 400 equiv) dropwise with stirring at 2O0C To this was added (2,4- dichlorophenyl)-N-methylmethanamme (880 mg, 4 63 mmol, 1 19 equiv) dropwise with stirring at 2O0C The resulting solution was stirred for 4 h at 2O0C The solids were filtered out The resulting mixture was concentrated under vacuum to give 1 5 g (84%) of N-(4-(2-((2,4-dichlorobenzyl)(methyl)ammo)acetyl)phenyl)acetamide as a white solid
Figure imgf000188_0002
Intermediate 30.5: N-(4-(2-((2,4-dichlorobenzyl)(methyI)amino)-l- hydroxyethyl)phenyl)acetamide: Into a 100-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of N-(4-(2- ((2,4-dichlorobenzyl)(methyl)amino)acetyl)phenyl)acetamide (1 5 g, 4 11 mmol, 1 00 equiv) in methanol (20 mL) This was followed by the addition of NaBHU (300 mg, 7 89 mmol, 2 06 equiv) in several batches at 0-50C The resulting solution was stirred for 2 h at 0-5°C The reaction was then quenched by the addition of 5 mL of acetone The resulting mixture was concentrated under vacuum The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 10-1 5) This resulted in 1 2 g (76%) of N-(4-(2-((2,4-dichlorobenzyl)(methyl)amino)- 1 -hydroxyethyl)phenyl)acetamide as yellow oil
Figure imgf000189_0001
Intermediate 30.6: N-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenyl)acetamide: Into a 100-mL 3-necked round-bottom flask, was placed a solution of N-(4-(2-((2,4-dichlorobenzyl)(methyl)amino)-l- hydroxyethyl)phenyl)acetamide (500 mg, 1 36 mmol, 1 00 equiv) in dichloromethane (3 mL) This was followed by the addition of sulfuric acid (3 mL) dropwise with stirring at O0C The resulting solution was stirred for 5 h at 0-50C The reaction was then quenched by the addition of 20 mL of water/ice The pH value of the solution was adjusted to 7-8 with sodium hydroxide The resulting solution was extracted with 3x20 mL of ethyl acetate and the organic layers combined and concentrated under vacuum The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 10-1 5) This resulted m 25 mg (5%) of N-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinohn-4-yl)phenyl)acetamide as a white solid 1H-NMR (300HMz, CDCl3, ppm) δ 7 46-7 49(2H, d, J=8 4Hz), 7 23-7 29(1H, m), 7 12-7 15(2H, d, J=8 4Hz), 6 80 (IH, s), 4 314(1H, s), 3 92(1H, d), 3 58-3 63(1H, d), 3 06(1H, s), 2 61- 2 68(1 H, m), 2 57(3H, s), 220(3 H, s) MS (ES, m/z) 349 [M+H]+
Figure imgf000190_0001
Intermediate 30.7: 4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)benzenamine: Into a 100-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of N-(4-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoqumolm-4-yl)phenyl)acetamide (2 g, 5 73 mmol, 1 00 equiv) in ethanol (20 mL) This was followed by the addition of sodium methanolate (5 g, 92 59 mmol, 16 16 equiv) in several batches, while the temperature was maintained at reflux The resulting solution was heated to reflux overnight The reaction was then quenched by the addition of 50 mL of water/ice The resulting solution was extracted with 3x50 mL of ethyl acetate and the organic layers combined and concentrated under vacuum This resulted in 1 5 g (85%) of 4-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoquinolin-4-yl)benzenamine as yellow oil 1H-NMR (300MHz, DMSO, ppni) δ 7 42-7 42(1H, d, J-I 5Hz), 6 83-6 86(2H, d, J=8 IHz), 6 78-6 78(1H, d, J=I 2Hz), 6 48-6 51(2H, d, J=8 4Hz), 4 98(2H, s), 4 02-4 O6(1H, m), 3 62 3 67(1H, d, J=162Hz), 3 43-3 48(1H, d, J=15 9Hz), 2 80-2 86(1 H, m), 2 37(3H, s) MS (ES, m/z) 307 [M+H]+
Figure imgf000190_0002
Intermediate 30.8: diethyl 2-(ch]orosulfonylamino)ethylphosphonate: Into a 100- mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of sulfuryl dichloπde (1 1 g, 8 15 mmol, 1 47 equiv) in dichloromethane (10 mL) This was followed by the addition of a solution of diethyl 2- ammoethylphosphonate (intermediate 1 9) (1 0 g, 5 52 mmol, 1 00 equiv) and tπethylamine (800 mg, 7 92 mmol, 1 43 eqmv) in dichloromethane (20 mL) dropwise with stirring at 00C The resulting solution was stirred for 2 h at O0C The reaction was then quenched by the addition of ice water The organic layer was washed with saturated sodium chloride (20 mL), dπed over anhydrous sodium sulfate and concentrated under vacuum This resulted in 0 5 g (crude) of the title compound as a colorless oil
Figure imgf000191_0001
Intermediate 30.9: diethyl 2-(N-(4-(6,8-dichIoro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenyl)sulfamoylamino)ethylphosphonate: Into a 50- mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed diethyl 2-(chlorosulfonylamino)ethylphosphonate (intermediate 30 8) (670 mg, 2 40 mmol, 1 47 equiv), 4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolm-4- yl)benzenamme (intermediate 30 7) (500 mg, 1 63 mmol, 1 00 equiv), N-ethyl-N- isopropylpropan-2-amine (400 mg, 3 10 mmol, 1 91 equiv) m acetomtnle (20 mL) The resulting solution was stirred for 3 h at 6O0C The resulting mixture was concentrated under vacuum and the residue was applied to a silica gel column and eluted with dichloromethane/methanol (20 1) This resulted in 150 mg (16%) of the title compound as a light yellow solid
Figure imgf000192_0001
Compound 30: 2-(N-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenyl)sulfamoylamino)ethylphosphonic acid: Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of diethyl 2-(N-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenyl)sulfamoylamino)ethylphosphonate (100 mg, 0 18 mmol, 1 00 equiv) in dichloromethane (5 mL) and bromotπmethylsilane (275 mg, 1 80 mmol, 9 89 equiv) The resulting solution was stirred overnight at 390C The resulting mixture was concentrated under vacuum and the residue was dissolved in dichloromethane (5 mL) This was followed by the addition of a solution of sodium hydroxide (14 5 mg, 0 36 mmol, 2 00 equiv) in methanol (02 mL) dropwise with stirring The solids were collected by filtration and dπed under reduced pressure This gave 40 mg (40%) of a sodium salt of the title compound as a white solid 1H-NMR (300MHz, J5-DMSO, pprri) δ 9 78 (IH, brs), 7 54 (IH, s), 7 47 (IH, brs), 7 09-7 17 (4H, m), 6 82 (IH, s ), 4 31 (IH, brs), 3 88 (2H, brs), 3 13 (IH, brs), 3 04 (2H, brs), 2 90 (IH, brs ), 2 58 (3H, s), 1 65-1 77 (2H, m) MS( m/z) 494 [M+H]+
Example 31
2-(N-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenyl)sulfamoylamino)ethylphosphonic acid
Figure imgf000192_0002
Intermediate 31.1: 2-bromo-l-(3-nitropheny])ethanone: Into a 500-mL 3-necked round-bottom flask, was placed a solution of l-(3-mtrophenyl)ethanone (50 g, 303 03 mmol, 1 00 eqmv) in acetic acid (300 mL), Br2 (53 5 g, 331 6 mmol, 1 00 equiv) The resulting solution was stirred for 2 h at 6O0C in an oil bath The reaction was then quenched by the addition of ice and the solids were collected by filtration The crude product was re-crystallized from ethyl acetate/petroleum ether in the ratio of 1 10 This resulted in 25 g (34%) of 2-bromo-l-(3-mtrophenyl)ethanone as a white solid
Figure imgf000193_0001
1 4-dioxane/Et3N
Figure imgf000193_0003
Figure imgf000193_0002
Intermediate 31.2: 2-((2,4-dichlorobenzyl)(methyl)amino)-l-(3- nitrophenyl)ethanone: Into a 100-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 2-bromo 1- (3-mtrophenyl)ethanone (2 g, 8 23 mmol, 1 00 equiv), tπethylamme (3 4 g, 4 00 equiv), (2,4 dichlorophenyl)-N methylmethanamine (1 9 g, 10 05 mmol, 1 20 equiv), 1,4- dioxane (50 mL) The resulting solution was stirred for 2 h at room temperature at which time it was judged to be complete by LCMS The mixture was concentrated under vacuum and the residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 100-1 50) This resulted in 1 5 g (50%) of 2-((2,4- dichlorobenzyl)(methyl)ammo)-l-(3-nitrophenyl)ethanone as a yellow solid
Figure imgf000193_0004
Intermediate 31.3: 2-((2,4-dichlorobenzyl)(methyl)amino)-l-(3- nitrophenyl)ethanol: Into a 500-mL 3-necked round-bottom flask, was placed a solution of 2-((2,4-dichlorobenzyl)(methyl)ammo)-l-(3-mtrophenyl)ethanone (28 g, 1 00 eqmv, Crude) in methanol (280 mL), NaBH4 (6 38 mg, 0 17 mmol, 200 equiv) The resulting solution was stirred for 0 5 h at O0C The reaction progress was monitored by LCMS The reaction was then quenched by the addition of 10 mL of acetone The resulting mixture was concentrated under vacuum The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 10—1 5) This resulted in 14 g of 2- ((2,4-dichlorobenzyl)(methyl)ammo)-l-(3-mtrophenyl)ethanol as a yellow solid
Figure imgf000194_0001
Intermediate 31.4: 6,8-dichIoro-2-methyl-4-(3-nitrophenyl)-l,2,3,4- tetrahydroisoquinoline: Into a 500-mL 3-necked round-bottom flask, was placed a solution of 2-((2,4-dichlorobenzyl)(methyl)ammo)-l-(3-mtrophenyl)ethanol (14 g, 39 55 mmol, 1 00 eqmv) in dichloromethane (140 mL), sulfuric acid (140 mL) The resulting solution was stirred overnight at room temperature The reaction progress was monitored by LCMS The resulting solution was diluted with 100 mL of ice The pH value of the solution was adjusted to 8-9 with sat sodium hydroxide (100 mL) The resulting solution was extracted with 2x500 mL of ethyl acetate and the organic layers combined and dried over sodium sulfate The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 10—1 5) This resulted m 7 g (51%) of 6,8- dichloro-2-methyl-4-(3-nitrophenyl)-l,2,3,4-tetrahydroisoquinoline as a yellow solid
Figure imgf000195_0001
Intermediate 31.5: 3-(6,8-dichloro-2-methyI-l,2,3,4-tetrahydroisoquinolin-4- yl)benzenamine: Into a 100-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed 6,8-dichloro-2-methyl-4-(3- nitrophenyl)-l,2,3,4-tetrahydroisoqmnolme (200 mg, 0 59 mmol, 1 00 equiv), Fe (360 mg, 6 43 mmol, 8 60 equiv), hydrogen chloπde (0 02 mL), ethanol (06 mL), water (0 2 mL) The resulting solution was stirred for 0 5 h at 8O0C in an oil bath The solids were filtered out The resulting mixture was concentrated under vacuum This resulted in 02 g (crude) of 3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin 4 yl)benzenamme as yellow oil
Figure imgf000195_0002
Compound 31: 2-(N-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquιnolin-4- yl)phenyl)sulfamoylamino)ethylphosphonic acid: Following the procedures outlined m Example 30, substituting 3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolm-4- yl)amline (intermediate 31 5) for 4-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoqumolm-4-yl)amlme gave the title compound as a sodium salt 1H- NMR(300MHz, D2O+DMSO-d6, ppm) h i 61 (s, IH), 7 33 (t, J=8 IHz, IH), 7 07-7 15 (m, 2H), 6 81-6 86 (m, 2H), 4 39 4 66 (m, 3H), 3 75 3 81 (m, IH), 3 45 3 50 (m, IH), 3 02-3 08 (m, 5H), 1 67-1 78 (m, 2H) MS (ES, m/z) 494 0 [M+H]+ Example 32
3-(N-(4-(6,8-dichloro-2-methyI-l,2,3,4-tetrahydroisoquinolin-4- yl)phenyl)sulfamoylamino)propylphosphonic acid
Figure imgf000196_0001
Compound 32: 3-(N-(4-(6,8-dichIoro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenyl)sulfamoylamino)propylphosphonic acid: Following the procedures outlined in Example 30, substituting 3-diethyl 3-aminopropylphosphonate 10 (intermediate 4.1) for diethyl 2-aminoethylphosphonate gave the title compound as a sodium salt. 1H-NMR (300MHz, CD3OD, ppm): δ 7.47(s, IH), 7.28(s, 4H), 6.81(s, IH), 4.73-4.77(m, 2H), 4.57(m, IH), 3.81(s, IH), 3.66(s, IH), 3.18(s, 3H), 3.06(s, 2H), 1.74(m, 4H), 1.20-1.35(m, IH). MS (ES, m/z): 508 [M+H]+
I5 Example 33
3-(N-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqninolin-4- yl)phenyl)sulfamoylamlno)propylphosphonic acid
Figure imgf000196_0002
20 Compound 33: 3-(N-(3-(6,8-dichloro-2-methyI-l,2,3,4-tetrahydroisoquinolin-4- yl)phenyl)sulfamoylamino)propylphosphonic acid: Following the procedures outlined in Example 30, substituting 3-diethyl 3-ammopropylphosphonate (intermediate 4 1) for diethyl 2-aminoethylphosphonate and 3-(6,8-dichloro-2-methyl- l,2,3,4-tetrahydroisoquinohn-4-yl)anilme (intermediate 31 5) for 4-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)anilme gave the title compound as a sodium salt 1H-NMR (300MHz, CD3OD, ppm) δ 7 54(s, IH), 7 38(s, IH), 7 25(s, IH), 7 l l(s, IH), 6 94(m, 2H), 4 66(s, IH), 4 55-4 51(m, IH), 3 89(s, IH), 3 65(m, 2H), 3 18(s, 3H), 3 05(s, 2H), 1 71(m, 4H) MS (ES, m/z) 508 [M+H]+
10
Example 34
(2S)-2-(3-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenyl)ureido)succinic acid
Figure imgf000197_0001
Intermediate 34.1: (2S)-dimethyl 2-(3-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenyl)ureido)succinate: Into a 50-mL 3-necked round- bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a
20 solution of 4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumohn-4-yl)benzenamine (intermediate 30 7) (200 mg, 0 65 mmol, 1 00 equiv) in dichloromethane (10 mL), tπethylamme(l 2 mL) This was followed by the addition of bis(tπchloromethyl) carbonate (200 mg, 0 67 mmol, 1 03 equiv) slowly with stirring at 0 50C The resulting solution was stirred for 1 h at room temperature To this was added tπethylamine (1
25 mL) followed by (S)-dimethyl 2-ammosuccinate (200 mg, 1 24 mmol, 1 91 equiv) m several batches The resulting solution was stirred for 2 h at room temperature The resulting mixture was concentrated under vacuum and the residue was applied onto a silica gel column and elrued with ethyl acetate/petroleum ether (1 10-1 5) This resulted in 50 mg (15%) of (2S)-dimethyl 2-(3-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolm-4-yl)phenyl)ureido)succinate as yellow oil
Figure imgf000198_0001
Compound 34: (2S)-2-(3-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin- 4-yl)pheny])ureido)succinic acid: Into a 50-mL round-bottom flask, was placed a solution of (2S)-dimethyl 2-(3-(4-(6,8-dichloro-2-methyl-l,253,4-tetrahydroisoqumolm- 4-yl)phenyl)ureido)succinate (100 mg, 0 20 mmol, 1 00 equiv) in methanol(5 mL), water (1 mL), sodium hydroxide (30 mg, 0 75 mmol, 3 71 equiv) The resulting solution was stirred for 3 h at room temperature and then concentrated under vacuum The pH of the solution was adjusted to 3-4 with IN hydrochloric acid The solids were collected by filtration and the residue was lyophihzed This resulted in 16 mg (16%) of (2S)-2-(3- (4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolin-4-yl)phenyl)ureido)succimc acid as a white solid 1H-NMR (300MHz, DMSO, ppm) δ 8 98(s, IH), 7 66(s, IH), 7 38-744(d, J=17 IHz, 2H), 7 12-7 15(d, J=8 4Hz, 2H), 6 78(s, IH), 6 60-6 63(s, IH), 448-4 54(m, 4H), 3 63-3 66(s, 2H), 3 01(s, IH), 2 51-2 84(m, 2H) MS (ES, m/z) 466 [M+H]+
Example 35
(2S)-2-(3-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenyl)ureido)succinic acid
Figure imgf000199_0001
Compound 35: (2S)-2-(3-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin- 4-y])phenyl)ureido)succinic acid: Following the procedures outlined in Example 34, substituting 3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolin-4-yl)anilme
(intermediate 31 5) for 4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumohn-4- yl)anilme gave, after purification by preparative HPLC, the title compound as a TFA salt 1H-NMR (300MHz, DMSO, ppm) δ 8 88(s, IH), 7 54(s,lH), 7 31-7 18(m, 3H), 6 83-6 78(m, 2H), 6 53-6 51 (m, IH), 4 49-4 47 (m, IH), 4 29(m, IH), 3 87(m, 2H),
10 3 32(m, 2H), 2 76-2 59(m, 2H), 2 50(s, 3H) MS 466 [M+H]+
Example 36
(2S)-2-(3-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinoIin-4- yl)phenyl)ureido)pentanedioic acid
I5
Figure imgf000199_0002
Compound 36: (2S)-2-(3-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin- 4-yl)phenyl)ureido)pentanedioic acid: Following the procedures outlined in
20 Example 34, substituting (S)-diethyl 2-aminopentanedioate for (S)-dimethyl 2- ammosuccmate gave the title compound Η-NMR(300MHz, DMSO, ppm) δ 12 32(s, 2H), 8 63(s, IH), 7 47(s, IH), 7 30-7 33(d, J=8 IHz, 2H), 7 06-7 09(d, J=5 4Hz, 2H), 6 79(s, IH), 645-648(d, J-8 IHz, IH), 4 19-420(s, 2H), 3 68(s, 2H), 2 95(s, IH), 2 68(s, IH), 2 45(s, 3H)1 2 27-2 30(s, 2H), 1 99-2 02(s, IH), 1 76-7 78(s, IH) MS (ES, m/z) 480 [M+H]+
Example 37
(2S)-2-(3-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinoIin-4- yl)phenyl)ureido)pentanedioic acid
Figure imgf000200_0001
10
Compound 37: (2S)-2-(3-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinoIin-
4-yl)phenyl)ureido)pentanedioic acid: Following the procedures outlined in Example 34, substituting 3-(6,8-dichloro 2-methyl-l,2,3,4-tetrahydroisoquinolm-4-yl)amline (intermediate 31 5) for 4-(6,8-dichloro-2-methyl-l,2,3,4 tetrahydroisoquinolin-4-
I5 yl)aniline and (S)-diethyl 2-ammopentanedioate for (S)-dimethyl 2-ammosuccmate gave, after purification by preparative HPLC, the title compound as a TFA salt 1H- NMR (300MHz, DMSO-^ , ppm) δ 8 74(s, IH), 7 67(s, IH), 7 42(m, IH), 7 27- 7 25(m, 2H), 6 79(m, 2H), 6 52-6 49(m, IH), 4 63-4 58(m, IH), 444(m, 2H), 4 20- 4 16(m, IH), 3 72-3 64(m, 2H), 2 99(s, 3H), 2 34-2 27(m, 2H), 2 01-1 97(m, 2H), 1 82-
20 1 77(m, 2H) MS 480 [M+H]+
Example 38
(3-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenyl)ureido)methylphosphonic acid
25
Figure imgf000201_0001
Intermediate 38.1: 4-nitrophenyl 4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylcarbamate: Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 4- (6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumohn-4-yl)benzenamine (intermediate 30 7) (300 mg, 0 98 mmol, 1 00 equiv) in dichloromethane (10 mL) This was followed by the addition of 4-nitrophenyl chloro formate (230 mg, 1 14 mmol, 1 20 equiv) in several batches at room temperature The resulting solution was stirred for 3 h at room
10 temperature The solids were collected by filtration This resulted in 0 3 g (65%) of 4- mtrophenyl 4-(6,8-dichloro-2-methyl- 1 ,2,3 ,4-tetrahydroisoquinolm-4- yl)phenylcarbamate as a yellow solid
Figure imgf000201_0002
I5
Intermediate 38.2: diethyl (3-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenyl)ureido)methyIphosphonate: Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 4-nitrophenyl 4-(6,8-dichloro-2-methyl-l, 2,3,4-
20 tetrahydroisoqumohn-4-yl)phenylcarbamate (200 mg, 042 mmol, 1 00 equiv) in N,N- dimethylformamide (6 mL), a solution of diethyl aminomethylphosphonate (144 mg, 0 63 mmol, 1 50 equiv) in N,N-dimethylformamide (1 mL) and tπethylamme (64 mg) The resulting solution was stirred overnight at room temperature The reaction was then quenched by the addition of 10 mL of water The resulting solution was extracted with 3x10 mL of ethyl acetate and the organic layers combined and dπed over anhydrous sodium sulfate and concentrated under vacuum This resulted in 40 mg (17%) of diethyl (3-(4-(6,8-dichloro-2-methyl- 1 ,2,3,4-tetrahydroisoqumohn-4- yl)phenyl)ureido)methylphosphonate as a solid
Figure imgf000202_0001
Compound 38: (3-(4-(6,8-dichIoro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenyl)ureido)methylphosphonic acid: Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of diethyl (3-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolin-4- yl)phenyl)ureido)methylphosphonate (40 mg, 008 mmol, 1 00 equiv) in dichloromethane (5 mL) and bromotπmethylsilane (0 15 mL) The resulting solution was stirred overnight at room temperature The resulting mixture was concentrated under vacuum To the above was added methanol (5 mL) and sodium hydroxide (5 mg) The resulting mixture was stirred 0 5 h at room temperature The solids were collected by filtration and the residue was lyophihzed This resulted in 17 4 mg (42%) a sodium salt of the title compound as a yellow solid 1H-NMR (300MHz, CD3OD+DC1 , ppm) δ 7 46-7 49(m, 3H), 7 20-7 23(d, J=S 7Hz, 2H), 6 80(s, IH), 4 77-4 83(d, J-15 9Hz, IH), 4 65-4 71(m, IH), 4 50-4 55(d, 7=16 2Hz, IH), 3 79-3 85(m, IH), 3 56-3 69(m, 3H), 3 32(s, 3H) MS (ES, m/z) 444 [M+H]+
Example 39 (3-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinoIin-4- yl)phenyl)ureido)methylphosphonic acid
Figure imgf000203_0001
Compound 39: (3-(3-(6,8-dichIoro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenyl)ureido)methylphosphonic acid: Following the procedures outlined in Example 38, substituting 3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolm-4- yl)amlme (intermediate 31 5) for 4-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoquinolin-4-yl)amlme gave the title compound as a sodium salt 1H- NMR(300MHz, CD3OD, ppm) δ 7 47 (s, IH), 7 37 (m, 3H), 6 96 (m, IH), 6 82 (s, IH), 4 81 (m, IH), 4 70 (m, IH), 4 54 (m, IH), 3 83 (m, IH), 3 65 (m, 3H), 3 19 (s, 3H)
Example 40
2-(3-(3-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenyl)ureido)propyl)malonic acid
Figure imgf000203_0002
Intermediate 40.1: ethyl 3-(3-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenyl)ureido)propanoate: Following the procedures outlined in Example 34, substituting ethyl 3-ammopropanoate for (S)-dimethyl 2- ammosuccmate gave the title compound as a yellow oil
Figure imgf000204_0001
Intermediate 40.2: 3-(3-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- y])phenyl)ureido)propanoic acid: Into a 50-mL round-bottom flask, was placed a solution of ethyl 3-(3-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolm-4- yl)phenyl)ureido)propanoate (150 mg, 0 33 mmol, 1 00 eqmv) m methanol (10 mL), water (2 mL) and sodium hydroxide (80 mg, 2 00 mmol) The resulting solution was stirred for 2 h at 250C and the resulting mixture was concentrated under vacuum The pH value of the solution was adjusted to 7 8 with hydrogen chloπde The resulting solution was extracted with chloroform (3x10 ml) and the organic layers combined and dπed over sodium sulfate This resulted in 31 5 mg (22%) of 3-(3-(4-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoqumolin-4-y])phenyl)ureido)ρropanoic acid as a white solid 1H-NMR (300MHz, DMSO, ppm) δ 8 56(1 H, s), 7 45(1H, s), 7 29-7 32(2H, d, J-8 IHz), 7 04-7 07(2H, d, J-8 4Hz), 6 79(1 H, s), 6 21(1H, s), 4 16(1H, m), 3 56- 3 58(2H, d, J-5 4Hz), 3 27-3 29(2H, d, J"6Hz), 2 82-2 87(1H, m), 2 59(2H, s), 2 38- 240(4H, m) MS (ES, m/z) 422 [M+H]+
Figure imgf000205_0001
Intermediate 40.3: l-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenyl)-3-(3-(2,2-dimethyI-4,6-dioxo-l,3-dioxan-5-yl)-3-oxopropyl)urea: Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 3-(3-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinohn- 4-yl)phenyl)ureido)propanoic acid (200 mg, 047 mmol, 1 00 equiv) m dichloromethane (20 mL), N-(3-dimethylaminopropyl)-N'-ethylcarbodnmide hydrochloπde (136 mg, 0 71 mmol, 1 50 equiv) and 4-dimethylammopyπdme (115 mg, 0 94 mmol, 1 99 equiv) This was followed by the addition of a solution of 2,2-dimethyl-l,3-dioxane-4,6-dione (102 mg, 0 71 mmol, 1 49 equiv) in dichloromethane (2 mL) dropwise with stirring at O0C The resulting solution was stirred for 3 h at room temperature The resulting mixture was washed with KHSO4 (2x10 mL) The mixture was dπed over anhydrous sodium sulfate and concentrated under vacuum This resulted m 240 mg (92%) of l-(4- (6,8-dichloro-2-memyl-l,2,3,4-tetrahydroisoqumolin-4-yl)phenyl)-3-(3-(2,2-dimethyl- 4,6-dioxo-l,3-dioxan-5-yl)-3-oxopropyl)urea as a yellow solid
Figure imgf000205_0002
Intermediate 40.4: l-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenyl)-3-(3-(2,2-dimethyl-4,6-dioxo-l,3-dioxan-5-yl)propyI)urea: Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of l-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenyl)-3-(3-(2,2-dimethyl-4,6-dioxo-l,3-dioxan-5-yl)-3-oxopropyl)urea (150 mg, 027 mmol, 1 00 eqmv) in dichloromethane(10 niL) and acetic acid (1 mL) Sodium borohydπde (42 mg, 1 11 mmol, 404 eqmv) was added and the resulting solution was stirred overnight at room temperature The resulting mixture was washed with saturated aqueous sodium chloride (3x10 mL) The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum This resulted in 30 mg (21%) of l-(4 (6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoqumohn-4-yl)phenyl)-3-(3-(2,2-dimethyl-4,6-
10 dioxo-l,3-dioxan-5-yl)propyl)urea as a yellow solid
Figure imgf000206_0001
Compound 40: 2-(3-(3-(4-(6,8-dichIoro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-
15 yl)phenyl)ureido)propyl)malonic acid: Into a 50-mL round-bottom flask, was placed a solution of l-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqmnolm-4-yl)phenyl)-3-
(3-(2,2-dimethyl-4,6-dioxo-l,3-dioxan-5-yl)propyl)urea (100 mg, 0 19 mmol, 1 00 equiv) m 2,2,2-tπfluoroacetic acid (10 mL), and water (2 mL) The resulting solution was stirred overnight at room temperature The resulting mixture was concentrated
20 under vacuum The residue was applied onto a silica gel column with methanol water
(60%) The residue was lyophihzed This resulted in 36 3 mg (30%) of a TFA salt of the title compound as a white solid 1H-NMR (300MHz, DMSO, ppm) δ 8 55(s, IH),
7 64(s, IH), 7 39-7 42(d, J=8 7Hz, 2H), 7 09-7 12(d, J-S 4Hz, 2H), 6 79(s, IH), 6 23-
6 27(m, IH), 4 33-4 50(m, 3H), 3 62(s, IH), 3 19(m, IH), 3 08-3 10(d, J=S 7Hz, 2H),
25 2 94(s, 3H), 1 70-1 77(d, /-23 IHz, 2H), 1 41-1 46(d, 7-12Hz, 2H) MS (ES, m/z)
494 [M+H]+ Example 41
NjN'-Cbutane-l^-diyObisKE^N^diaminomethyleneVS-CS^-difluoro-^C^ sulfamoylphenoxy)phenyl)-2-methylacrylamide]
Figure imgf000207_0001
Intermediate 41.1 (E)-ethyl 2-methyl-3-(3,4,5-trifluorophenyl)acrylate: To a solution of dry DMF (5OmL) under N2 was added 3,4,5-tπfluorobenzaldehyde (4 26g,
10 26 6mmol) followed by ethyl 2-(tπphenylphosphoranybdene)propionate (10 6g,
29 3mmol) in portions, keeping the solution at room temperature After 1 hour, TLC
(10% EtOAC in Hexanes) showed complete conversion, and the solvent was removed by rotary evaporation The resulting material was brought up in 5OmL methyl t-butyl ether (MBTE) and the precipitate removed by filtration and washed with additional
I5 MBTE (3x50mL) After concentration, the resulting filtrate was applied onto a silica gel column (25% EtOAc m hexanes) resulting in 6 Og of the title compound (93%) as a white powder
Figure imgf000207_0002
20
Intermediate 41.2 (E)-ethyl 3-(3,5-difluoro-4-phenoxyphenyl)-2-methylacryIate:
To a solution of (E)-ethyl 2-methyl-3-(3,4,5-tπfluorophenyl)acrylate (Intermediate 41 1, 6 Og, 24 56mmol) in dry DMF (25mL) under N2 was added phenol (2 774g, 29 5mmol) and K2CO3 (10 2g, 73 68mmol) The resulting solution was brought to 25 120°C and stirred for 3 hours at which point TLC indicated complete conversion The solvent was removed by rotary evaporation and the resulting residue brought up in EtOAc (20OmL) and washed with water (2x200mL), IN NaOH (2x200mL) and bπne (20OmL) The organic layer was dπed over Na2SC>4 and concentrated to yield 6 94g (89%) of the title compound as tan crystals
Figure imgf000208_0001
Intermediate 41.3 (E)-ethyl 3-(4-(4-(chlorosulfonyl)phenoxy)-3,5-difluorophenyl)- 2-methylacrylate: To a solution of (E)-ethyl 3-(3,5-difluoro-4-phenoxyphenyl)-2- methylacrylate (intermediated 2) (Ig, 3 14mmol) m DCM (3 14mL) under N2 was added chlorosulfonic acid (O 419mL, 6 28mmol) dropwise After 1 hour an additional
IQ 0209mL chlorosulfonic acid was added After an additional hour the reaction mixture was quenched with ice-water and extracted into EtOAc (2x200mL) The combined organic layers were dπed briefly (<10min) over Na2SO4 and concentrated to recover
1 283g of the title compound (98%) as a yellow oil
Figure imgf000208_0002
Intermediate 41.4 N,N'-(butane-l,4-diyl)bis[4-(2,6-difIuoro-4-(2- carboethoxypropenyl)phenoxy)benzenesulfonamide]: To a solution of (E)-ethyl 3- (4-(4-(chlorosulfonyl)phenoxy)-3,5-difluorophenyl)-2-methylacrylate (Intermediate 20 41 3) (104 3mg, 025mmol) in chloroform (O 5mL) was added DIEA (O 0869mL, 0 5mmol) and a solution of butane- 1 ,4-diamine (12 6uL, 0 125mmol) and DIEA (0 087mL, 0 5mmol) in chloroform (0 125mL) After one hour the solvent was removed and the resulting residue brought up in EtOAc (4OmL), washed with water (2x40mL), bnne (4OmL) and dπed over Na2SC>4 Removing the solvent gave 118mg of the title compound which was used without farther purification
Figure imgf000209_0001
Intermediate 41.5 : N,N'-(butane-l,4-diyl)bis [4-(2,6-difluoro-4-(2- carboxypropenyl)phenoxy)benzenesulfonamide]: To a solution of Intermediate 41 4 (118mg, 0 139mmol) in MeOH (1 39mL) was added a NaOH (O 3M in water, 0 278mL, 0 835mmol) The reaction was placed under N2 and heated at 60°C for 30 minutes After cooling the reaction mixture was diluted with water (2OmL), partitioned with EtOAc (2OmL) and acidified with HCl After extracting with EtOAc (2x20mL) the combined organic phases were dπed over Na2SO4 and the solvent removed to give 40 7mg of the title compound
Figure imgf000209_0002
Compound 41: N,N'-(butane-l,4-diyl)bis[(E)-N-(diaminomethylene)-3-(3,5- difluoro-4-(4-sulfamoyIphenoxy)phenyl)-2-methylacrylamide]: Thionyl chloride (2 mL) was added to intermediate 41 5 (40 7 mg, 0 051 mmol) and was heated at 80c under N2 After 70 minutes, the solvent was removed in vacuo The residue was brought up in toluene (2mL) and the toluene was also removed in vacuo The bis-acid chloπde was dissolved in DME (0 5 mL) and added to guamdine free base (1 4 mmol, prepared as follows To a slurry of guamdine hydrochloπde (480 mg, 5 0 mmol) was added 25% NaOMe in MeOH (1 03 mL, 4 5 mmol) The mixture was stirred for 30 minutes and then filtered A portion of the filtrate (0 40 mL) was concentrated to dryness ) m DME (ImL) After 15 minutes, water (10 mL) was added and the mixture was extracted with EtOAc (3 x 25 mL) The organic layer was dπed (Nβ24) and concentrated The crude product was purified by preparative HPLC to give the title compound (7 8 mg) as the TFA salt 1H-NMR (400 mHz, CD3OD) δ 7 80 (d, 4H), 7 44 (s, 2H), 7 30 (d, 4H), 7 11 (d, 4H), 2 80 (m, 4H), 2 18 (s, 6H), 1 44 (m, 4H) MS (m/z) 875 16 (M+H)
Example 42 N,N'-(l,4-phenylenebis(methylene))bis[(E)-N-(diaminomethylene)-3-(3,5-difluoro- 4-(4-sulfamoylphenoxy)phenyl)-2-methylacrylamide]
Figure imgf000210_0001
Compound 42: N,N'-(l,4-phenyIenebis(methylene))bis[(E)-N-(diaminomethylene)- 3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-2-methylacrylamide]): Following the procedures outlined in Example 41, compound 42 was made using 1,4- phenylenedimethanamine as the amine Purification by preparative HPLC gave the title compound as a TFA salt 1H-NMR (400 mHz, CD3OD) δ 7 87 (d, 4H), 7 44 (s, 2H), 7.31 (d, 4H), 7.06 (d, 6H), 7.04 (s, 2H), 4.02 (s, 4H), 2.19 (s, 6H) MS (m/z): 92421 (M+H)
Example 43
N,N'-(2,2'-(ethane-l,2-diylbis(oxy))bis(ethane-2,l-diyl))bis[(E)-N-
(diaminomethylene)-3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-2- methylacrylamide]
Figure imgf000211_0001
Intermediate 43.1 N,N'-(2,2'-(ethane-l,2-diylbis(oxy))bis(ethane-2,l-diyl))bis((E)- 4-(2,6-difluoro-4-(2-carboethoxypropenyl)phenoxy)benzenesulfonaraide): To a solution of (E)-ethyl 3-(4-(4-(chlorosulfonyl)phenoxy)-3,5-difluorophenyl)-2- methylacrylate(mtermediate 41.3) (225 mg, 0 54 mmol) m DCM (3 mL) was added a solution of 2,2'-(ethane-l,2-diylbis(oxy))diethanarmne (38 mg, 0 26 mmol) and tπethylamine (101 mg, 1 0 mmol) m DCM (2 mL) dropwise After 30 minutes, IN HCl was added (10 mL) and the reaction mixture was extracted with DCM (3 x 15 mL) The combined organic layers were dπed (Na2SC>4) and concentrated to give the title compound (262 mg)
Figure imgf000211_0002
Intermedeate 43.2 N,N'-(2,2'-(ethane-l,2-diylbis(oxy))bis(ethane-2,l-diyl))bis((E)- 4-(2,6-difluoro-4-(2-carboxypropenyl)phenoxy)benzenesulfonamide): A solution of the intermediate 43 1 (262 mg, 029 mmol) and 3N NaOH (0 6 mL, 1 8 mmol) in methanol (3 mL) was heated at 650C for 1 hour The reaction mixture was cooled to RT and the methanol removed at reduced pressure and IN HCl (3 mL, 3 mmol) was added to the residue The product was extracted into DCM (3 x 15 mL) The combined organic layers were dried (Na2SO4) and concentrated to give the title compound (173 mg)
Figure imgf000212_0001
Compound 43: N,N'-(2,2'-(ethane-l,2-diylbis(oxy))bis(ethane-2,l-diyl))bis[(E)-N- (diaminomethylene)-3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-2- methylacrylamide]: Thionyl chloπde (1 mL) was added to intermediate 43 2 (63 mg, 0 074 mmol) and was heated at 80° After 2 hours, the solvent was removed in vacuo The bis-acid chloπde was dissolved in DME (1 mL) and added to guanidine free base (1 4 mmol, prepared as follows To a slurry of guanidine hydrochloride (480 mg, 5 0 mmol) was added 25% NaOMe in MeOH (1 03 mL, 4 5 mmol) The mixture was stirred for 30 minutes and then filtered A portion of the filtrate (040 mL) was concentrated to dryness ) in DME (ImL) After 15 minutes, water (10 mL) was added and the mixture was extracted with EtOAc (3 x 25 mL) The organic layer was dried (Na2SO4) and concentrated The crude product was purified by preparative HPLC to give the title compound (20 mg) as the TFA salt 1H-NMR (400 mHz, CD3OD) δ 7 83 (d, j = 8 8 Hz, 4H), 7 43 (s, 2H), 7 30 (d, j = 8 9 Hz, 4H), 7 11 (d, j-8 6 Hz, 4H), 3 42 (t, j=5 5 Hz, 8H), 3 03 (t, j=5 4 Hz, 4H), 2 17 (s, 6H) MS (m/z) 935 08 (M+H)
Example 44 N,N'-(2,2'-(2,2'-oxybis(ethane-2,l-diyl)bis(oxy))bis(ethane-2,l-diyl))bis[(E)-N-
(diaminomethylene)-3-(3,5-difluoro-4-(4-sulfamoyIphenoxy)phenyl)-2- methylacrylamide]
Figure imgf000213_0001
Intermediate 44.1: (E)-ethyl 3-(4-(4-(N-(2-(2-(2-(2- azidoethoxy)ethoxy)ethoxy)ethyl)sulfamoyl)phenoxy)-3,5-difluorophenyl)-2- raethylacrylate- To a solution of (E)-ethyl 3-(4-(4-(chlorosulfonyl)phenoxy)-3,5- difluorophenyl)-2-methylacrylate (intermediate 41 3) (250 mg, 0 60 mmol) in DCM (3 mL) was added a solution of 2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethanamme (157 mg, 0.72 mmol) and tπethylamine (72 mg, 0.72 mmol) in DCM (2 mL). After 15 minutes, water (10 mL) was added and the reaction mixture was extracted with DCM (2 x 25 mL) The combined orgaic layers were washed with water (10 mL), bπne (10 mL), dπed (Na2SO4) and concentrated. The crude material was purified by flash chromatography on silica gel eluting with 50% EtOAc in DCM to give the title compound (169 mg)
Figure imgf000213_0002
Intermediate 44.2: (E)-ethyl 3-(4-(4-(N-(2-(2-(2-(2- aminoethoxy)ethoxy)ethoxy)ethyl)sulfamoyl)phenoxy)-3,5-difluorophenyI)-2- methylacrylate: To a solution of (E)-ethyl 3-(4-(4-(N-(2-(2-(2-(2- azidoethoxy)ethoxy)ethoxy)ethyl)sulfamoyl)phenoxy)-3,5-difluorophenyl)-2- methylacrylate (169 mg, 028 mmol) in THF (6 ml) and water (O 6 mL) under nitrogen was added tnmethylphosphme (26 mg, 0 34 mmol) After stirring for 3 hours, the solvents were removed at reduced pressure and The residue was dissolved in water (5 mL) and extracted with EtOAc (3 x 25 mL) The combined organic layers were dried (Na2SO4) and concentrated to give the title compound (162 mg)
Figure imgf000214_0001
Intermediate 44.3: N,N'-(2,2'-(2,2'-oxybis(ethane-2,l-diyl)bis(oxy))bis(ethane-2,l- diyl))bis[4-(2,6-difluoro-4-(2-carboethoxypropenyl)phenoxy)benzenesulfonamide]:
A solution of (E)-ethyl 3-(4-(4-(chlorosulfonyl)phenoxy)-3,5-difluorophenyl)-2- methylacrylate (mtrmediate 41 3) (71 mg, 0 17 mmol) in EtOAc (1 mL) was added to a solution of (E)-ethyl 3-(4-(4-(N-(2-(2-(2 (2- ammoethoxy)ethoxy)ethoxy)ethyl)sulfamoy])phenoxy)-3,5-difluorophenyl)-2- methylacrylate (84 mg, 0 15 mmol) and tnethylamine (22 mg, 022 mmol) in DCM (1 mL) with stirring After 30 minutes, water (10 mL) was added and the product extracted into DCM (3 x 15 mL) The combined organic layers were dried (NaSC^) and concentrated to give the title compound (177 mg)
Figure imgf000214_0002
Compound 44 N,N'-(2,2'-(2,2'-oxybis(ethane-2,l-diyI)bis(oxy))bis(ethane-2,l- diyl))bis[(E)-N-(diaminomethylene)-3-(3,5-difluoro-4-(4- sulfamoylphenoxy)phenyl)-2-methylacryIamide] Following the procedures outlined in Example 43, intermediate 44 3 was converted to the bis-guamdine and gave, after 5 purification by preparative HPLC, the title compound (21 mg) as a TFA salt 1H-NMR (400 mHz, CD3OD) δ 7 84 (d, j = 8 8 Hz, 4H), 744 (s, 2H), 7 30 (d, j = 8 8 Hz, 4H), 7 10 (d, j=8 8 Hz, 4H), 3 54 (m, 4H), 3 48 (m, 4H), 3 43 (t, j=5 5 Hz, 4H), 3 04 (t, j=5 5 Hz, 4H), 2 17 (d, j=l 2 Hz, 6H) MS (m/z) 979 05 (M+H)
10 Example 45
(E)-3-(4-(4-(N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)sulfamoyl)phenoxy)- 3,5-difluorophenyl)-N-(diaminomethylene)-2-methylacrylamide
Figure imgf000215_0001
I5
Compound 45: (E)-3-(4-(4-(N-(2-(2-(2-(2- aminoethoxy)ethoxy)ethoxy)ethyl)sulfamoyl)phenoxy)-3,5-difluorophenyl)-N- (diaminomethylene)-2-methylacrylamide: A 4 3 M solution of guanidme free base in methanol was prepared A 25% solution of NaOMe in MeOH (1 03 mL, 4 5 mmol)
20 was added to guanidine hydrochlonde (480 mg, 5 0 mmol), and the mixture was stirred for 30 minutes The mixture was filtered (0 2 μ, PTFE) to give the guanidme free base solution A portion (03 mL, 1 3 mmol) was added to (E) ethyl 3 (4 (4-(N-(2-(2-(2-(2- ammoethoxy)ethoxy)ethoxy)ethyl)sulfamoyl)phenoxy)-3,5-difluorophenyl)-2- methylacrylate (74 mg, 0 13 mmol) with stirring After 15 minutes, water (10 mL) was
25 added and the product extracted with DCM (4 x 20 mL) The combined organic layers were dπed (Na2SU4) and concentrated The crude product was purified by preparative HPLC to give the title compound (34 mg) as a TFA salt 1H-NMR (400 mHz, d6 DMSO) δ 11 14 (s, IH), 8 38 (br s, 4H), 7 78 (d, j = 9 0 Hz, 2H), 7 5 (m, 3H), 7 45 (d, ]=9 1, 2H), 7 42 (s, IH), 7 19 (d, j=8 8 Hz, 2H), 3 55 (m, 6H), 3 44 (m, 4H), 3 36 (m, 2H), 2 95 (m, 2H), 2 87 (m, 2H), 2 11 (s, 3H) MS (m/z) 586 11 (M+H)
Example 46
N,N'-(13-oxo-3,6,9,17,20,23-hexaoxa-12,14-diazapentacosane-l,25-diyI)bis[(E)-N-
(diaminomethylene)-3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-2- methylacrylamide]
Figure imgf000216_0001
Intermediate 46.1 N,N'-(13-oxo-3,6,9,17,20,23-hexaoxa-12,14-diazapentacosane- l,25-diyl)bis[4-(2,6-difluoro-4-(2- carboethoxypropenyl)phenoxy)benzenesulfonamide]: Carbonyldiimidisole (16 2 mg, 0 10 mmol) was added to a solution of (E)-ethyl 3-(4-(4-(N-(2-(2-(2-(2- aminoethoxy)ethoxy)ethoxy)ethyl)sulfamoyl)phenoxy) 3,5-difluorophenyl)-2- methylacrylate ( intermediate 44 2) (125 mg, 0 22 mmol) in DMF (2 mL) and stirred for 23 hours at which time the solvent was removed under vacuum The residue was dissolved in EtOAc, washed with water (4 x 10 mL), dπed (Na2SO<ι) and concentrated to give the title compound (132 mg)
Figure imgf000217_0001
Compound 46: N,N'-(13-oxo-3,6,9,17,20,23-hexaoxa-12,14-diazapentacosane-l,25- diyI)bis[(E)-N-(diaminomethylene)-3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)- 2-methyIacrylamide] : A solution of 4 4 M guamdme in methanol (Example 45) (0 5 mL, 2 2 mmol) was added to a solution of intermediate 46 1 (65 mg, 0 055 mmol) in DMF, and stirred for 4 hours The reaction was quenched with 50% aqueous AcOH, and then concentrated to dryness The residue was purified by preparative HPLC to give the title compound (35 mg) as a TFA salt 1H-NMR (400 mHz, CD3OD) δ 7 84 (d, j = 8 2 Hz, 4H), 7 43 (d, j=l 4 Hz, 2H), 7 30 (d, j = 9 0 Hz, 4H), 7 11 (d, j=9 0 Hz, 4H), 3 57(m, 12H), 3 46 (m, 12H), 3 26 (t, J=5 4 Hz, 4H), 3 04 (t, j=5 4 Hz, 4H), 2 17 (d, j=l 3 Hz, 6H) MS (m/z) 1197 07 (M+H)
Example 47 N,N'-(13,20 dioxo-3, 6, 9, 24, 27, 30-hexaoxa-12, 21-diazadotricontane-l,32- diyl)bis[(E)-N-(diaminomethylene)-3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-
2-methylacryIamide]
Figure imgf000218_0001
Compound 47: N,N'-(13,20 dioxo-3, 6, 9, 24, 27, 30-hexaoxa-12, 21- diazadotricontane-l,32-diyl)bis[(E)-N-(diaminomethylene)-3-(3,5-difluoro-4-(4- sulfamoylphenoxy)phenyl)-2-methylacrylamide]: Following the procedures m Example 46, substituting subaπc acid bis(N-hydroxysuccimmide ester) for carbonyldiimidazole gave the title compound as a TFA salt H-NMR (400 mHz, CD3OD) δ 7 84 (m, , 4H), 743 (m, 2H), 7 30 (m, 4H), 7 11 (m, 4H), 3 58 (m, 12H), 3 50 (m, 8H), 3 32 (m, 4H), 3 05 (t, j=5 4 Hz, 4H), 2 18 (d, j=l 6 Hz, 6H), 2 15 (m, 4H),
10 1 56 (m, 4H)5 1 29 (m, 4H) MS (Wz) 1309 12 (M+H)
Example 48
(E)-N-(diaminomethylene)-3-(3,5-difluoro-4-(4-(N-(2-(2-(2-(2-(4-(hydroxymethyl)- lH-l,2,3-triazol-l-yl)ethoxy)ethoxy)ethoxy)ethyI)sulfamoyl)phenoxy)phenyl)-2- 15 methylacrylamide
Figure imgf000219_0001
Intermediate 48.1: (E)-3-(4-(4-(N-(2-(2-(2-(2- azidoethoxy)ethoxy)ethoxy)ethyl)sulfamoyl)phenoxy)-3,5-difluorophenyl)-N- (diaminomethylene)-2-methylacrylamide: To (E)-ethyl 3-(4-(4-(N-(2-(2-(2-(2- azidoethoxy)ethoxy)ethoxy)ethyl)sulfamoyl)phenoxy)-3,5-difluorophenyl)-2- methylacrylate (250 mg, 0 42 mmol) was added 44 M guamdine m in methanol (as prepared m example 45) (1 0 mL, 44 mmol) and the reaction was stirred at RT After 30 minutes, water (10 mL) was added, and the mixture was extracted with DCM (4 x 25 mL) The aqueous phase was adjusted to pH 7, and extracted with DCM (2 x 25 mL) The combined organic extracts were dπed (Na2SO4) and concentrated to give the title compound (245 mg)
Figure imgf000219_0002
Compound 48: (E)-N-(diaminomethylene)-3-(3,5-difluoro-4-(4-(N-(2-(2-(2-(2-(4- (hydroxyniethyl)-lH-l,2,3-triazoI-l- yl)ethoxy)ethoxy)ethoxy)ethyl)sulfamoyl)phenoxy)phenyl)-2-methylacrylamide:
To a mixture of (E)-3-(4-(4-(N-(2-(2-(2-(2- azidoethoxy)ethoxy)ethoxy)ethyl)sulfamoyl)phenoxy)-3,5-difluorophenyl)-N- (diaminomethylene)-2-methylacrylamide (70 mg, O i l mmol) and propargyl alcohol 5 (64 mg, 0 11 mmol) in t-butanol (022 mL) and water (022 mL) was added 1 M sodium ascorbate (11 μL, 0 011 mmol) and 0 3 M copper sulfate (3 6 μL, 0 0011 mmol) and the reaction was stirred at RT After 14 hours, the product was purified by preparative HPLC to give the title compound (22 mg) as a TFA salt 1H-NMR (400 mHz, CD3OD) δ 7 93 (s, IH), 7 84 (m, , 2H), 7 44 (s, IH), 7 30 (m, 2H), 7 11 (m, 2H), 10 4 64 (d, j=0 6 Hz, 2H), 4 55 (t, j=5 0 Hz, 2H), 3 86 (t, j=5 0 Hz, 2H), 3 57 (m, 4H), 3 52-3 42 (m, 6H), 3 03 (t, j=5 4 Hz, 2H), 2 18 (d, j=l 3 Hz, 3H) MS (m/z) 668 14 (M+H)
Example 49
15 N,N'-(2,2'-(2,2'-(2,21-(2,2'-(4,4'-oxybis(methyIene)bis(lH-l,2,3-triazo]e-4,l- diyl))bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l- diyI))bis(oxy)bis(ethane-2,l-diyl))bis[(E)-N-(diaminomethylene)-3-(3,5-difluoro-4- (4-su]famoylphenoxy)phenyl)-2-methylacrylanude]
Figure imgf000220_0001
Compound 49: N,N1-(2,2'-(2,2'-(2,2'-(2,2'-(4,41-oxybis(methylene)bis(lH-l,2,3- triazole-4,l-diyl))bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l- diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis[(E)-N- 25 (diaminomethylene)-3-(3,5-difluoro-4-(4-sulfamoyIphenoxy)phenyl)-2- methylacrylamide] : Following the procedures in example 48, substituting propargyl ether for propargyl alcohol gave the title compound as a TFA salt 1H-NMR (400 mHz, CD3OD) δ 8 00 (s, 2H), 7 83 (m, 4H), 7 43 (s, 2H), 7 30 (m, 4H), 7 10 (m, 4H), 4 61 (s, 4H), 4 55 (m, 4H), 3 86 (m, 4H), 3 58-3 50 (m, 8H), 3 50-3 40 (m, 12H), 3 01 (m, 4H), 2 17 (d,j=1 3 Hz, 6H) MS (m/z) 1317 09 (M+H)
Example 50
N,N'-(2,2'-(piperazine-l,4-diyl)bis(ethane-2,l-diyl))di-((E)-N-(diaminomethylene)- 3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-2-methylacrylamide)
Figure imgf000221_0001
Intermediate 50.1: 2,2'-(piperazine-l,4-diyl)diacetonitrile. To a solution of piperazme (6 g, 69 77 mmol, 1 00 equiv) in acetomtπle (150 mL) was added potassium carbonate (19 2 g, 139 13 mmol, 2 00 equiv) and the mixture was stirred To this was added dropwise a solution of 2-bromoacetomtπle (16 7 g, 140 34 mmol, 2 00 equiv) in acetomtπle (100 mL) and the suspension was stirred for 4 h at room temperature The solids were filtered out and the resulting solution was concentrated under vacuum The crude product was purified by re-crystallization from methanol resulting in 7 75 g (68%) of Intermediate 50 1 as a white solid
,C'"~"'N''~xi L1AIH4/THF H2N—\ / — \ N'' I J. ^c -,-N ^N N^
-NH2
Intermediate 50.2: 2,2'-(piperazine-l,4-diyl)diethanamine. To a suspension of lithium aluminum hydπde (L1AIH4, 700 mg, 18 42 mmol, 4 30 equiv) in tetrahydrofuran (40 mL) cooled to 00C was added dropwise a solution of Intermediate 50 1 (700 mg, 427 mmol, 1 00 equiv) in tetrahydrofuran (10 mL) The mixture was stirred for 15 minutes at O0C and heated to reflux for 3 h The reaction was cooled, the pH adjusted to 8-9 with potassium hydroxide (50%), and the solids filtered out The resulting mixture was concentrated under vacuum and the resulting solids washed with hexane to afford 0 3 g (41%) of Intermediate 502 as a yellow solid
Figure imgf000222_0001
Intermediate 50.3: N,N'-(2,2'-(piperazine-l,4-diyl)bis(ethane-2,l-diyl))bis(4- (benzyloxy)benzenesulfonamide). To Intermediate 502 (500 mg, 2 91 mmol, 1 00 equiv) m dichloromethane (10 mL) was added tnethylamme (1 46 g, 0 01 mmol, 2 00 equiv) and 4-(benzyloxy)benzene-l-sulfonyl chloπde (2 0 g, 0 01 mmol, 240 equiv) and the resulting solution was stirred for 2 h at room temperature The reaction was diluted with dichloromethane, washed with 3x10 mL of water, dπed over sodium sulfate then filtered and concentrated under vacuum to afford 0 9 g (47%) of Intermediate 50 3 as a yellow solid
Figure imgf000222_0002
Intermediate 50.4: N,N'-(2,2'-(piperazine-l,4-diyl)bis(ethane-2,l-diyl))bis(4- hydroxybenzenesulfonamide). To intermediate 50 3 (3 g, 4 52 mmol, 1 00 equiv) in N,N-dimethylformamide (500 mL) and methanol (100 mL) was added Palladium on carbon (1 g) and the suspension stirred under hydrogen gas for 4 h at room temperature The solids were filtered out and the resulting mixture was concentrated under vacuum to afford 1 5 g (69%) of Intermediate 504 as a gray solid
Figure imgf000223_0001
Intermediate 50.5: N,N'-(2,2'-(piperazine-l,4-diyl)bis(ethane-2,l-diyI))bis((E)- ethyl 3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-2-methylacrylate) To
Intermediate 504 (1 g, 2 06 mmol, 1 00 eqmv) in N1N dimethylformamide (30 mL) was added CS2CO3 (1 45 g, 445 mmol, 2 16 equiv) and the resulting suspension stirred for 2 h at room temperature To this was added a solution of (E)-ethyl 2-methyl-3- (3,4,5-tπfluorophenyl)acrylate (intermediate 41 1) (1 1 g, 4 51 mmol, 2 19 equiv) in N,N-dimethylformamide (10 mL) dropwise with stirring The reaction was stirred for
10 05 h at room temperature and then overnight at 9O0C The resulting mixture was concentrated under vacuum, the residue was applied onto a silica gel column and then eluted with dichloromethane methanol (100 1) to afford 390 mg (20%) of Intermediate 50 5 as a yellow solid
Figure imgf000223_0002
I5 Intermediate 50.6: N,N'-(2,2'-(piperazine-l,4-diyl)bis(ethane-2,l-diyI))di-((E)-3- (3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-2-methylacrylic acid). To
Intermediate 50 5 (170 mg, 0 16 mmol, 1 00 eqmv, 90%) in 1 1 methanol / tetrahydrofuran (20 mL) was added lithium hydroxide (4 equiv, 30 mg) and the reaction was stirred for 2 h at 270C The pH value of the solution was adjusted to 1~2 with aqueous hydrochloric acid (6 mol/L) and the solids were collected by filtration The residue was washed with ethyl acetate(2x5 mL) and then dπed under vacuum to afford 150 mg (94%) of Intermediate 506 as a white solid
Figure imgf000224_0001
Compound 50: N,N'-(2,2'-(piperazine-l,4-diyl)bis(ethane-2,l-diyl))di-((E)-N- (diaminomethylene)-3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-2- methylacrylamide): To a solution of Intermediate 50 6 (100 mg, 0 09 mmol, 1 00 equiv, 80%) in tetrahydrofuran (30 mL) was added carbonyl diimidazole (CDI, 58 mg, 0 36 mmol, 4 00 equiv) and the resulting solution was stirred for 1 h at 250C To this was added guamdine (2M in methanol, 10 ml) and the resulting solution was stirred for an additional 14 h at 3O0C The resulting mixture was concentrated under vacuum, the residue was applied onto a silica gel column and eluted with dichloromethane methanol (10 1) The crude product (230 mg) was then puπfied by reverse-phase (Cl 8) preparative-HPLC to afford 16 mg (17%) of a formate salt of the title compound as a white solid 1H-NMR (300MHz,CD3OD, ppm) 1 89-7 92(4H,d, J-8 7Hz), 7 50(2H,s), 7 34-7 36(4H,d,J=8 7Hz), 7 16-7 19(4H,d,J=8 7Hz), 2 88-3 16(16H,m), 2 20(6H,s), MS (ES, m/z) 959 [M+H]+ Example 51
(E)-4-(4-(4-(3-(diaminomethyleneamino)-2-methyl-3-oxoprop-l-enyl)-2,6- difluorophenoxy)phenylsulfonamido)phenylphosphonic acid
Figure imgf000225_0001
Intermediate 51.1: (E)-3-(3,5-difluoro-4-phenoxyphenyl)-2-methylacrylic acid. To a solution of (E)-ethyl 3-(3,5-difluoro-4-phenoxyphenyl)-2-methylacrylate (intermediate 41 2) (900 mg, 2 83 mmol, 1 00 equiv) in methanol (20 mL) was added methanolic 2M LiOH (50 mL) and the resulting solution stirred for 2 h The resulting mixture was concentrated under vacuum, the pH value of the solution was adjusted to 5-6 with aqueous HCl (6 mol/L) and the mixture was extracted with 3x20 mL of ethyl acetate The organic layers were combined, washed with 2x10 mL of sodium chloride (sat ) and then dπed over anhydrous sodium sulfate The solids were filtered out and the solution was concentrated to afford 0 7 g (85%) of Intermediate 51 1 as a white solid
Figure imgf000225_0002
Intermediate 51.2: (E)-3-(4-(4-(chlorosulfonyl)phenoxy)-3,5-difluorophenyl)-2- methylacrylic acid. To Intermediate 51 1 (1 g, 3 14 mmol, 1 00 equiv) in dichloromethane (15 mL) at 0-50C was added dropwise a solution of sulfurochloπdic acid (8 5 g, 73 28 mmol, 23 00 equiv) in dichloromethane (5 mL) The reaction was stirred overnight at 250C in an oil bath, and then quenched by the addition of 200 mL of water/ice The mixture was extracted with 4x50 mL of dichloromethane and the organic layers combined and dπed over anhydrous sodium sulfate to afford 1 1 g (90%) of Intermediate 51 2 as a yellow solid
Figure imgf000226_0001
Intermediate 51.3: (E)-3-(4-(4-(N-(4-
(diethoxyphosphoryI)phenyI)suIfainoyI)phenoxy)-3,5-difluorophenyl)-2- methylacrylic acid To diethyl 4-ammophenylphosphonate (intermediate 2 2) (150 mg, 0 66 mmol, 1 00 eqmv) m pyridine (3 mL) was added Intermediate 51 2 (300 mg, 0 77 mmol, 1 22 eqmv) in several portions The mixture was stirred for 3 h at 3O0C and then concentrated, the pH value of the solution adjusted to 3 with aqueous HCl (1 mol/L) and the resulting mixture extracted with 3x30 mL of ethyl acetate The organic layers were combined, dried over anhydrous sodium sulfate, concentrated, applied onto a silica gel column and eluted with dichloromethane methanol (50 1) to afford 100 mg (26%) of Intermediate 51 3 as a yellowish solid
Figure imgf000226_0002
Intermediate 51.4: (E)-diethyl 4-(4-(4-(3-(diaminomethyleneamino)-2-methyl-3- oxoprop-l-enyl)-2,6-difluorophenoxy)phenylsuIfonamido)phenylphosphonate. To Intermediate 51 3 (150 mg, 0 26 mmol, 1 00 equiv) in tetrahydrofuran (2 mL) was added CDI (120 mg, 0 74 mmol, 1 40 eqmv) and the reaction stirred for 2 h at RT To this was added guanidme (IM in DMF, 0 8 ml) and the reaction was stirred overnight at 3O0C The resulting mixture was concentrated under vacuum and the crude product was purified by reverse phase (C18) Prep HPLC to afford 40 mg (25%) of Intermediate 51 4 as a White solid
Figure imgf000227_0001
Compound 51: (E)-4-(4-(4-(3-(diaminomethyleneamino)-2-methyl-3-oxoprop-l- enyl)-2,6-difluorophenoxy)phenylsulfonamido)phenylphosphonic acid: To
Intermediate 51 4 (40 mg, 0 06 mmol, 1 00 eqmv) in tetrahydrofuran (2 mL) was added bromotnmethylsilane (15 mg, 0 09 mmol, 1 37 equiv) dropwise with stirring and the resulting solution was stirred at 4O0C overnight The resulting mixture was concentrated, diluted with methanol (2 mL) and then concentrated under vacuum This operation was repeated four times The crude product (75 mg) was purified by reverse phase (C 18) Prep-HPLC to afford 12 5 mg of a formate salt of the title compound as a white solid 1H-NMR (300 MHz, DMSO, ppm) 10 54(s, IH), 7 82-7 79(d, J=8 4Hz, 2H), 7 52-7 40(m, 5H), 7 18-7 10(m, 4H), 2 08(s, 3H), 31P-NMR (400 MHz, DMSO, ppm) 11 29, MS (ES, m/z) 567 [M +H]+
Example 52
(E)-4-((4-(4-(3-(diaminoniethyleneainino)-2-methyl-3-oxoprop-l-enyl)-2,6- difluojrophenoxy)phenylsulfOnamido)methyl)benzyIphosphonic acid
Figure imgf000227_0002
Intermediate 52.1: diethyl 4-((4-(benzyloxy)phenylsulfonamido)methyl)benzyl- phosphonate. To 4-diethyl 4-(aminomethyl)benzylphosphonate (intermediate 6 1) (60 mg, 0 23 mmol, 1 00 equiv) in dichloromethane (10 mL), tπethylamme (47 mg, 0 47 mmol, 2 00 equiv) was added dropwise a solution of 4-(benzyloxy)benzene-l-sulfonyl chloride (72 mg, 026 mmol, 1 10 equiv) in dichloromethane (5 mL) and the resulting solution was stirred for 1 h at 250C The reaction mixture was concentrated, the residue applied onto a silica gel column and then eluted with ethyl acetate/petroleum ether (1 1) The isolated product was washed with 2x50 mL of n-hexane resulting in 50 mg (43%) of Intermediate52 1 as a white solid
Figure imgf000228_0001
Intermediate 52.2: diethyl 4-((4-hydroxypheny]sulfonamido)methyl)benzy]- phosphonate To Intermediate 52 1 (1 2 g, 2 39 mmol, 1 00 eqmv) in methanol (20 mL) in N,N-dimethylformamide (5 mL) was added Palladium on carbon (0 9 g) and the suspension stirred overnight at 3O0C under a hydrogen atmostphere The reaction was filtered and concentrated under vacuum to afford 1 g (91%) of Intermediate 522 as brown oil
Figure imgf000228_0002
Intermediate 52.3: (E)-ethyl 3-(4-(4-(N-(4-((diethoxyphosphoryl)methyl)benzyl)- sulfamoyI)phenoxy)-3,5-difluorophenyI)-2-methylacrylate. To Intermediate 52 2 (100 mg, 024 mmol, 1 00 equiv) m N,N-dimethylformamide (10 mL) was added CS2CO3 (160 mg, 049 mmol, 2 10 equiv) and the mixture was stirred for 1 5 h at room temperature To this was added a solution of (E)-ethyl 2-methyl-3-(3,4,5- tπfluorophenyl)acrylate (intermediate 41 1) (60 mg, 0 25 mmol, 1 10 equiv) in N,N- dimethylformamide (5 mL) and the reaction was stirred overnight at 900C The solids were filtered out and the filtrate was concentrated under vacuum, the residue applied onto a silica gel column and eluted with dichloromethane / methanol (200 1) to afford 50 mg (23%) of Intermediate 52 3 as yellow oil
220
Figure imgf000229_0001
Intermediate 52.4: (E)-3-(4-(4-(N-(4-
((diethoxyphosphoryl)methyI)benzyl)sulfaπioyl)-phenoxy)-3,5-difluorophenyl)-2- methylacrylic acid. To Intermediate 52 3 (700 mg, 1 10 mmol, 1 00 equiv) in tetrahydrofuran (20 mL) and water (20 mL) was added LiOH (700 mg, 29 17 mmol, 30 00 equiv) and the resulting solution was stirred for 1 h at 25°C The reaction was concentrated, the pH value of the solution was adjusted to 4-5 with aqueous HCl (2 mol/L) and the mixture was extracted with 2x150 mL of ethyl acetate The organic
10 layers were combined, dπed over anhydrous sodium sulfate, concentrated, the residue applied onto a silica gel column and then eluted with ethyl acetate/petroleum ether (1 1- 2 1) to afford 250 mg (35%) of Intermediate 52 4 as a white solid
Figure imgf000229_0002
I5
Compound 52: (E)-4-((4-(4-(3-(diaminomethyleneamino)-2-methyl-3-oxoprop-l- enyl)-2,6-difluorophenoxy)phenylsu)fonamido)methyl)benzyIphosphonic acid.
Compound 52 was prepared from Intermediate 52 4 using the procedures descπbed under Example 51, except preparative HPLC was not required, affording 84 mg (89%)
20 of a white solid , 1H-NMR (300MHz, CD3OD, ppm) 7 83-7 80(d, J-% 7Hz, 2H), 7 52(s, IH), 7 38-7 36(d, 7=8 7Hz, 2H), 723-7 20(m, 2H), 7 17-7 09(m, 4H), 4 06(s, 2H), 3 1 l(s, IH), 3 04(s, IH), 2 23 2 23(s, 3H) MS (ES, m/z) 595 [M+H]+
Example 53 (E)-4-(4-(4-(3-(diaminomethyleneamino)-2-methyl-3-oxoprop-l-enyl)-2,6- difluorophenoxy)phenylsulfonamido)benzylphosphonic acid
Figure imgf000230_0001
Compound 53 : (E)-4-(4-(4-(3-(diaminomethyleneamino)-2-methyl-3-oxoprop-l- enyl)-2,6-difluorophenoxy)phenylsulfonamido)benzylphosphonic acid. Compound 53 was prepared from diethyl 4-aminobenzylphosphonate (intermediate 3.2) using the procedures described in Example 52 except the final product was purified by preparative HPLC. 1H-NMR (300 MHz, CD3OD, ppm): 7.77-7.74(d, J=8.7Hz, 2H), 7.46(s, IH), 7.33-7.31(d, J=8.7Hz, 2H), 7.21-7.19(m, 2H), 7.06-7.1 l(m, 4H), 3.04- 2.97(d, 7=21.6Hz, 2H), 2.19(s, 3H); 31P-NMR (400 MHz, CD3OD, ppm): 22.49. MS (ES, m/z):581 [M+H]+.
Example 54
(E)-3-(4-(4-(3-(diaminomethyleneamino)-2-methyl-3-oxoprop-l-enyI)-2,6- difluorophenoxy)phenylsulfonamido)propylphosphonic acid
Figure imgf000230_0002
Compound 54: (E)-3-(4-(4-(3-(diaminomethyleneamino)-2-methyl-3-oxoprop-l- enyl)-2,6-difluorophenoxy)phenylsulfonamido)propylphosphonic acid. Compound 54 was prepared from diethyl 3-aminopropylphosphonate (intermediate 4.1) using the procedures described under Example 51. 1H-NMR (400 MHz, DMSO, ppm): 7.81- 7.78(d, J=8.4Hz, 2H), 7.57(s, IH), 7.42-7.39(d, J=9.3Hz, 2H), 7.22-7.19(d, J=8.7Hz, 2H), 2.75-2.77(q, 2H), 2.10(s, 3H), 1.59-1.42(m, 4H). MS (ES, m/z): 533 [M+H]+ Example 55
(E)-2-(4-(4-(3-(diaminomethyleneamlno)-2-methyl-3-oxoprop-l-enyl)-2,6- difluorophenoxy)phenylsulfonamido)ethylphosphonic acid
Figure imgf000231_0001
Compound 55: (E)-2-(4-(4-(3-(diaminomethyleneamino)-2-methyl-3-oxoprop-l- eny])-2,6-difluorophenoxy)phenylsulfonamido)ethylphosphonic acid Compound 55 was prepared from diethyl 2-aminoethylphosphonate (intermediate 1 9) using the
10 procedures described under Example 51, except purification of the final product by preparative HPLC was not required , Η-NMR(400MHz, DMSO, ppm) 11 02(s, IH), 828(s, 4H), 7 79-7 82(d, J=92Hz, 2H), 7 62-765(t, IH), 7 54-749(m, 3H), 726- 7 24(d, J=H 8Hz, 2H), 3 42 3 58(m, 2H), 2 15(s, 3H), 1 73-1 65(m, 2H), 31P-NMR (400MHz, DMSO, ppm) 21 36 MS (ES, m/z) 519 [M+H]+
I5
Example 56
(E)-(4-(4-(3-(diaminomethyleneamino)-2-methyl-3-oxoprop-l-enyl)-2,6- difluorophenoxy)phenylsulfonamido)methylphosphonic acid
Figure imgf000231_0002
Compound 56: (E)-(4-(4-(3-(diaminomethyleneamino)-2-methyl-3-oxoprop-l- enyl)-2,6-difluorophenoxy)phenylsulfonamido)methylphosphonic acid Compound 56 was prepared from diethyl aminomethylphosphonate (intermediate 5 3) using the 25 procedures descπbed under Example 51, except purification of the final product by Flash-Prep-HPLC with CH3CN water (10 100) 1H -NMR(300MHz, DMSO, ppm) δ 7 84-7 81(d, J-8 1Hz, 2H ), 7 57(s, IH), 7 45-7 42(d, J=9 3Hz, 3H ), 7 18-7 15(d, J-8 4Hz, 2H), 3 04-3 01 (m, 2H), 2 08(s, 3H) MS (ES, m/z) 505 [M+H]+
Example 57
(E)-2-(4-(4-(3-(diaminomethyleneamino)-2-methyl-3-oxoprop-l-enyl)-2,6- difluorophenoxy)-N-(phosphonomethyl)phenylsulfonamido)acetic acid
Figure imgf000232_0001
Compound 57: (E)-2-(4-(4-(3-(diaminomethyleneamino)-2-methyl-3-oxoprop-l- enyl)-2,6-difluorophenoxy)-N-(phosphonomethyl)phenyl-sulfonamido)acetic acid
Compound 57 was prepared from ethyl 2-((diethoxyphosphoryl)methylammo)acetate (intermediate 8 2) using the procedures described under Example 51 1H-NMR (300MHz, DMSO, ppm) δ 8 33(s, 4H), 7 84-7 81(d, J-8 IHz, 2H), 7 52-7 50(d, J-7 8Hz, 2H), 7 19-7 16(d, J-8 4Hz, 2H), 4 1 l(s, 2H), 2 14(s, 3H), MS (ES, m/z) 563 [M+H]+
Example 58
(E)-N-(diaminomethylene)-3-(3,5-difluoro-4-(4-(N-(2- methoxyethylcarbamoyl)sulfamoyl)phenoxy)phenyl)-2-methylacrylamide
Figure imgf000232_0002
Intermediate 58.1 : (E)-3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-2- methylacrylic acid. (E) 3-(4-(4-(chlorosulfonyl)phenoxy)-3,5-difluorophenyl)-2- methylacryhc acid (Intermediate 51 2) was converted to intermediate 58 1 using procedures outlined in Example 58, with aqueous ammonia as the amine The title compound was obtained as a yellow solid
Figure imgf000233_0001
Intermediate 58.2: (E)-methyl 3-(3,5-difluoro-4-(4-sulfamoylphenoxy)phenyl)-2- methylacrylate. Into a 50-mL round-bottom flask, was placed a solution of intermediate 58 1 (2 g, 5 42 rπmol, 1 00 equiv) in methanol (60 mL) This was followed by the addition of thionyl chlonde (2 5 g, 21 19 mmol, 4 00 equiv) dropwise with stirring at O0C The resulting solution was stirred for 3 h at 500C The resulting mixture was concentrated under vacuum The pH value of the solution was adjusted to 7 with ammonia (2 mol/L) The resulting solution was extracted with 10 mL of ethyl acetate and the organic layers combined and dried over anhydrous sodium sulfate and concentrated under vacuum The residue was applied onto a silica gel column with petroleum ether/ethyl acetate (30 1-1 1) This resulted in 2 1 g (97%) of the title compound as a white solid
Figure imgf000233_0002
Intermediate 58.3: (E)-methyl 3-(4-(4-(N-(ethoxycarbonyI)sulfamoyI)phenoxy)- 3,5-difluorophenyI)-2-methylacrylate. Into a 50-mL round-bottom flask, was placed a solution of intermediate 58 2 (280 mg, 0 73 mmol, 1 00 equiv) in acetone (20 mL) This was followed by the addition of potassium carbonate (200 mg, 1 45 mmol, 2 00 equiv) The mixture was stirred for 3 h at room temperature To this was added ethyl chloroformate (90 mg, 0 83 mmol, 1 20 equiv) The resulting solution was stirred for 6 h at 650C The resulting mixture was concentrated under vacuum The pH value of the solution was adjusted to 2-3 with hydrogen chlonde (1 mol/L) The resulting solution was extracted with 2x50 ml of ethyl acetate and the organic layers combined The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum This resulted in 300 mg (72%) of the title compund as yellow oil
Figure imgf000234_0001
Intermediate 58.4: (E)-methyl 3-(3,5-difluoro-4-(4-(N-(2-methoxyethylcarbamoyl)- sulfamoyl)phenoxy)phenyl)-2-methylacrylate Into a 100-mL round-bottom flask, was placed a solution of intermediate 58 3 (300 mg, 0 66 mmol, 1 00 equiv) in toluene (20 mL), 2-methoxyethanamine (100 mg, 1 33 mmol, 1 10 equiv) The resulting solution was stirred for 1 h at 1100C The resulting mixture was concentrated under vacuum The residue was applied onto a silica gel column with petroleum ether/ethyl
10 acetate (1 1) This resulted m 0 3 g (92%) of the title compound as a yellow solid
Figure imgf000234_0002
Compound 58: (E)-N-(diaminomethylene)-3-(3,5-difluoro-4-(4-(N-(2-
15 methoxyethylcarbamoyl)sulfamoyl)phenoxy)phenyI)-2-methylacrylamide
Intermediate 584 was converted to compound 58 using the procedures descπbed under Example 52 Purification by preparative HPLC gave a TFA salt of the title compound 1H-NMR (300MHz, DMSO, ppm) δlO 62(s, IH), 8 33 (s, 3H),7 94-7 91(d, J=S 7Hz, 2H), 7 55-7 52(d, J=9Hz, 2H), 7 45(s, IH), 7 26 7 22(d, J=9Hz, 2H), 6 55(s, IH), 3 37- 20 3 27(m, 2H), 3 21 (s, 3H), 3 15-3 12(m, 2H), 2 16(s, 3H) MS (ES, m/z) 512 [M+H]+
Example 59
(E)-2-(4-(4-(3-(diaminomethyleneamino)-2-methyl-3-oxoprop-l-enyl)-2,6- difluorophenoxy)phenylsulfonamido)succinic acid
25
Figure imgf000235_0001
Intermediate 59.1: (E)-di-tert-butyl 2-(4-(4-(3-(diaminomethyleneamino)-2- methyl-3-oxoprop-l-enyl)-2,6-difluorophenoxy)phenylsulfonatnido)succinate. Intermediate 59 1 was prepared from di-tert-butyl 2-ammosuccmate using the procedures descπbed under Example 51
Figure imgf000235_0002
Compound 59: (E)-2-(4-(4-(3-(diaminomethyleneamino)-2-methyl-3-oxoprop-l- enyl)-2,6-difluorophenoxy)phenylsulfonamido)succinic acid. Into a 50-mL round- bottom flask, was placed a solution of intermediate 59 1 (100 rag, 0 16 mmol, 1 00 eqmv) in tetrahydrofuran (5 mL) This was followed by the addition of 2,2,2- tπfluoroacetic acid (10 mL) dropwise with stirring The resulting solution was stirred for 3 h at room temperature The resulting mixture was concentrated under vacuum This resulted in 63 6 mg (64%) of a TFA salt of the title compound as a light yellow solid 1H-NMR (300MHz, DMSO, pprή) δ 8 26(s, 4H), 7 82-7 79(d, J=Z 7Hz, 2H), 7 49-7 45(m, 3H), 7 19-7 16(d, J=8 4Hz, 2H), 4 00-3 96(m, IH), 2 65-2 60(m, IH), 2 48-2 41(m, IH), 2 13(s, 3H) MS (ES, ra/z) 527 [M+H]+
Example 60
4-(3-(6-chloro-2-(diaminomethyleneamino)quinazolin-4-yl)phenyl)piperazine-l- carboximidamide
Figure imgf000236_0001
Intermediate 60.1: tert-butyl 4-(3-bromophenyl)piperazine-l-carboxylate: Into a 250-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed copper(I) iodide (1 0 g, 5 26 mmol, 0 20 equiv), L-prolme (930 mg, 8 09 mmol, 0 30 equiv) in DMSO (50 mL) The resulting solution was stirred for 15 mm at room temperature Then, tert-butyl piperazme-1-carboxylate (5 g, 26 88 mmol, 1 00 equiv), 1,3-dibromobenzene (9 5 g, 40 25 mmol, 1 50 equiv), potassium carbonate (74 g, 53 62 mmol, 1 99 equiv) was added The resulting solution was stirred overnight at 9O0C The reaction was then quenched by the addition of 100 mL of water The resulting solution was extracted with 2x100 mL of ethyl acetate and the organic layers combined and dried over anhydrous sodium sulfate and concentrated under vacuum The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 6) This resulted in 2 9 g of tert-butyl 4-(3-bromophenyl)piperazine-l- carboxylate as a white solid
Figure imgf000236_0002
Intermediate 60.2: 3-(4-(tert-butoxycarbonyl)piperazin-l-yl)phenylboronic acid: Into a 250-mL 3 -necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of tert-butyl 4-(3- bromophenyl)piperazme-l -carboxylate (3 8 g, 11 14 mmol, 1 00 equiv) m toluene/tetrahydrofuran=l 1 (40 mL) This was followed by the addition of n BuLi (4 9 mL, 2 5M/L) dropwise with stirring at -7O0C The resulting solution was stirred for 30 mm at 7O0C To this was added tπisopropyl borate (2 5 g, 13 30 mmol, 1 19 equiv)dropwise with stirring at -7O0C The mixture was warmed to O0C, the reaction was then quenched by the addition of 13 mL of saturated ammonium chloπde and 3 4 mL of water Phosphoric acid (85 wt %,1 5g,l 2 equiv) was added and the mixture was stirred for 30 min The organic layer was separated and dπed over anhydrous sodium sulfate and concentrated under vacuum The residue was dissolved in 20 mL of toluene The product was precipitated by the addition of 80 mL of heptane The solids were washed with 20 mL of heptane and collected by filtration This resulted in 2 9 g (85%) of 3-(4-(tert-butoxycarbonyl)piperazin-l-yl)phenylboromc acid as a white solid
Figure imgf000237_0001
Intermediate 60.3: 6-chloroquinazoline-2,4(lH,3H)-dione: Mo a 500-mL 3-necked round-bottom flask, was placed a solution of 2-ammo-5-chlorobenzoic acid (10 g, 58 48 mmol, 1 00 equiv) in water (100 mL), acetic acid (8 g, 133 33 mmol, 2 24 equiv) This was followed by the addition of NaOCN (8 2 g, 126 15 mmol, 2 13 equiv) The mixture was stirred for 30 rains at 3O0C To this was added sodium hydroxide (86 g, 2 15 mol, 37 00 equiv) The resulting solution was stirred overnight at 30cC The solids were collected by filtration The residue was dissolved in water The pH value of the solution was adjusted to 7 with hydrogen chloπde (12 mol/L) The solids were collected by filtration This resulted in 5 g (44%) of 6 chloroqmnazolme-2,4(lH,3H)-dione as a white solid
Figure imgf000237_0002
Intermediate 60.4 2,4,6-trichloroquinazoline: Into a 50-mL round-bottom flask, was placed a solution of 6-chloroquinazohne-2,4(lH,3H)-dione (2 2 g, 11 22 mmol, 1 00 equiv) ui 1,4-dioxane (20 mL), phosphoryl tπchlonde (17 g, 111 84 mmol, 1000 equiv) The resulting solution was stirred overnight at 120°C m an oil bath The resulting mixture was concentrated under vacuum The reaction was then quenched by the addition of 200 mL of water The resulting solution was extracted with 3x200 mL of ethyl acetate and the organic layers combined The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 50) This resulted in 1 8 g (69%) of 2,4,6- tπchloroqumazohne as a white solid
PdCI2(ClPPf)
Figure imgf000238_0002
Figure imgf000238_0001
Intermediate 60.5: tert-butyl 4-(3-(2,6-dichloroquinazolin-4-y])phenyl)piperazine- 1-carboxylate: Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 3-(4-(tert-butoxycarbonyl)piperazm-l- yl)phenylboromc acid (intermediate 602) (960 mg, 3 14 mmol, 1 00 eqmv) , 2,4,6- tπchloroqumazolme (800 mg, 3 43 mmol, 1 09 equiv), PdCl2(dppf) CH2Cl2 (130 mg, 0 16 mmol, 0 05 equiv), Potassium Carbonate (860 mg, 6 23 mmol, 1 99 equiv)m N,N- dimethylformamide (30 mL) The resulting solution was stirred for 3 h at 850C The reaction was then quenched by the addition of 50 mL of saturated brine The resulting solution was extracted with 2x30 mL of ethyl acetate and the organic layers combined and dπed over anhydrous sodium sulfate and concentrated under vacuum The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 6) This resulted m 0 45 g (31%) of tert-butyl 4-(3-(2,6-dichloroquinazolm-4- yl)phenyl)piperazme 1 carboxylate as a yellow solid
Figure imgf000238_0003
Intermediate 60.6: 2,6-dichloro-4-(3-(piperazin-l-yl)phenyl)quinazoline 2,2,2- trifluoroacetate. To intermediate 60 5 (100 mg, 0 22 mmol, 1 00 equiv) was added dichloromethane (10 mL) and 2,2,2-tπfluoroacetic acid (124 mg, 1 09 mmol, 5 00 equiv) and the resulting solution was stirred for 3 h at 4O0C The reaction was then concentrated under vacuum to afford 70 mg of Intermediate 60 6 as yellow solid
Figure imgf000239_0001
Intermediate 60.7: tert-butyl (4-(3-(2,6-dichloroquinazolin-4-yl)phenyl)piperazin- l-yl)methanediylidenedicarbamate To Intermediate 60 6 (70 mg, 0 15 mmol, 1 00 equiv) in dichloromethane (10 mL) was added N-tert-butoxycarbonyl-N'-tert- butoxycarbonyl-N"-tnfluoromethanesulfonylguamdine (91 mg, 0 23 mmol, 1 57 equiv) and tnethylamme (38 mg, 0 38 mmol, 2 54 equiv) and the resulting solution was stirred for 3 h at 4O0C The mixture was then concentrated under vacuum, the residue applied onto a silica gel column and eluted with ethyl acetate/petroleum ether (1 8) to afford 70 mg (77%) of Intermediate 60 7 as a yellow solid
Figure imgf000239_0002
Intermediate 60.8: tert-butyl (4-(3-(6-chloro-2-
(diaminomethyleneamino)quinazolin-4-yl)phenyl)piperazin-l- yl)methanediylidenedicarbamate To Intermediate 60 7 (70 mg, 0 12 mmol, 1 00 equiv) in NMP (1 5 mL) was added guamdine (0 24 mL, 2 00 equiv, 1 mol/L) and 1,4- diaza-bicyclo[2 2 2]octane (26 mg, 023 mmol, 1 99 equiv) and the resulting solution stirred for 1 5 h at 25°C The reaction was quenched by the addition of 20 mL of water and the resulting solution was extracted with 2x20 mL of ethyl acetate The organic layers were combined, dπed over anhydrous sodium sulfate, concentrated, the residue applied onto a silica gel column and eluted with dichloromethane/methanol (5 1) to afford 30 mg (41%) of Intermediate 60 8 as a yellow solid
Figure imgf000240_0001
Compound 60: 4-(3-(6-chloro-2-(dianύnomethyleneamino)quinazohn-4- yl)phenyl)piperazine-l-carboximidamide To Intermediate 60 8 (30 mg, 0 05 mmol, 1 00 equiv) in dichloromethane (5 mL) was added 2,2,2-tπfluoroacetic acid (0 2 mL) and the resulting solution stirred for 6 h at 30αC The mixture was then concentrated under vacuum and the residue lyophihzed to afford 20 mg (75%) of a TFA salt of the title compound as an off-white solid 1H-NMR (300 MHz, CD3OD, ppm) 7 97-8 08 (m, 3H), 7 54-7 59 (m, IH), 7 28-7 39 (m, 3H), 3 71 (d, J=4 8 Hz, 4H) , 3 44 (d, J=4 8 Hz, 4H) MS (ES, m/z) 424 0 [M+H]+
Example 61
2-(4-(4-(6-chloro-2-(diaminomethyleneamino)quinazolin-4-yl)phenyl)piperazin-l- yl)acetic acid
Figure imgf000241_0001
Intermediate 61.1 : 2,6-dichloro-4-(4-(piperazin-l-yl)phenyl)quinazoline hydrochloride. Following the procedures outlined in example 60, substituting 1,4- dibromobenzene for 1,3-dibromobenzene, 2,6-dichloro-4-(4-(prperazin-l- yl)phenyl)qumazolme hydrochloπde was obtained as a red solid
Figure imgf000241_0002
Intermediate 61.2: methyl 2-(4-(4-(2,6-dichIoroquinazolin-4-yl)phenyl)piperazin- l-yl)acetate. To methyl 2-bromoacetate (116 mg, 0 76 mmol, 3 00 equiv) in N,N- dimethylformamide (10 mL) was added potassium carbonate (140 mg, 1 01 mmol, 4 00 equiv) followed by the portion-wise addition of Intermediate 61 1 (100 mg, 0 25 mmol, 1 00 equiv) and the reaction was stirred for 4 h at 3O0C The mixture was concentrated under vacuum and the residue applied onto a silica gel column, elutmg with ethyl acetate/petroleum ether (1 5) to afford 60 mg (55%) of Intermediate 61 2 as a yellow solid
Figure imgf000242_0001
Intermediate 61.3: methyl 2-(4-(4-(6-chloro-2-
(diaminomethyleneamino)quinazoIin-4-yl)phenyl)piperazin-l-yl)acetate To
Intermediate 61 2 (60 mg, 0 14 mmol, 1 OO equiv) in NMP (5 mL) was added 1,4-diaza- bicyclo[2 22]octane (DABCO, 15 mg, O 13 mmol, 1 OO equiv), guamdme (O 3 mL of a IM solution m NMP, 2 00 equiv) and the resulting solution was stirred for 2 h at 300C The reaction was diluted with 10 mL of water, extracted with 4x10 mL of ethyl acetate and the organic layers combined and dπed over anhydrous sodium sulfate and then
10 concentrated under vacuum The residue was applied onto a silica gel column and eluted with dichloromethane/methanol (50 1-20 1) to afford 30 mg (47%) of Intermediate 61 3 as a yellow solid
Figure imgf000242_0002
I5
Compound 61 : 2-(4-(4-(6-chloro-2-(diaminomethyleneamino)quinazolin-4- yl)phenyl)piperazin-l-yl)acetic acid To Intermediate 61 3 (20 mg, 004 mmol, 1 00 equiv) in methanol (5 mL) was added a solution of LiOH (32 mg, 1 33 mmol, 30 00 equiv) in water (1 mL) and the reaction was stirred for 3 h at 250C The solution was concentrated under vacuum, the pH value adjusted to 6 with aqueous HCl (1 mol/L) and the resulting solids were collected by filtration to afford 15.6 mg (80%) of compound 61 as a yellow solid. 1H-NMR (300 MHz, DMSO pprri): 8.07- 8.06(t, IH), 7.96-7.93(t, 2H), 7.72-7.69(d, /=8.7Hz, 2H), 7.22-7.19(d, /=8.7Hz, 2H), 3.58-3.54(m, 4H), 3.43- 5 3.36(m, 6H). MS (ES, m/z): 440 [M+H]+.
Example 62
2-(4-(3-(6-chloro-2-(diaininomethyIeneamino)quinazolin-4-yl)phenyl)piperazin-l- yl)acetic acid
Figure imgf000243_0001
Compound 62: 2-(4-(3-(6-chloro-2-(diamiπomethyleneamino)quinazoIin-4- yl)phenyl)piperazin-l-yl)acetic acid. Compound 62 was prepared from intermediate 15 60.6, using the procedures described for Example 61. 1H-NMR (300 HHz, DMSOd6, ppm): 7.80-7.86 (m, 3H), 7.41-7.46 (m, IH), 7.16-7.22 (m, 2H), 7.08-7.10 (m, IH), 3.13 (brs, 4H), 2.71 (brs, 4H). MS (ES, m/z): 440 [M+H]+;
Example 63
20 2-(6-chloro-4-(3-(4-((2R,3S,4R,5R)-2,3,4,5,6-pentahydroxyhexanoyl)piperazin-l- yl)phenyl)quinazolin-2-yl)guanidine
Ac Ac
NaO °H °H ZnClj/AcjO/HCI O O
Na°rrYOH - HCX Λ Z ^Ac
O OH OH O ό O
Ac Ac Intermediate 63.1: (2R,3S,4R,5R)-2,3,4,5,6-pentaacetoxyhexanoic acid: Into a 50- mL 3-necked round-bottom flask, was placed ZnC^ (0 5 g, 0 50 equiv), acetic anhydπde(5 mL) To the above was added sodium (2S,3R,4S,5R)-2,3,4,5,6- pentahydroxyhexanoate (1 6 g, 6 97 mmol, 1 00 equiv, 95%) at -50C Anhydrous HCl was introduced m for 05 h at O0C The resulting solution was stirred overnight at room temperature The reaction mixture was cooled to O0C The reaction was then quenched by the addition of 8 g of ice The mixture was stirred for 1 h at room temperature The resulting solution was diluted with 20 mL of water The resulting solution was extracted with 3x20 mL of dichloromethane and the organic layers combined and dried over anhydrous sodium sulfate and concentrated under vacuum This resulted in 1 0 g (35%) of (2R,3S,4R,5R)-2,3,4,5,6-pentaacetoxyhexanoic acid as a yellow liquid
Figure imgf000244_0001
Intermediate 63.2: (2R,3R,4S,5R)-6-chloro-6-oxohexane-l,2,3,4,5-pentayl pentaacetate: Into a 50-mL 3-necked round-bottom flask, was placed a solution of (2R,3S,4R,5R)-2,3,4,5,6-pentaacetoxyhexanoic acid (intermediate 63 1) (610 mg, 1 35 mmol, 1 00 equiv, 90%) in CCl4 (30 mL) This was followed by the addition of oxalyl dichlonde (3 mL) dropwise with stirring The resulting solution was heated to reflux for 3 h in an oil bath The resulting mixture was concentrated under vacuum This resulted in 0 62 g (crude) of intermediate 63 2 as yellow oil
Figure imgf000244_0002
Intermediate 63.3: 2-(6-chloro-4-(3-(4-((2R,3S,4R,5R)-2,3,4,5,6- pentahydroxyhexanoyl)piperazin-l-yl)phenyl)quinazolin-2-yl)guanidine 2,2,2- trifluoroacetate To Intermediate 60 6 (150 mg, 0 32 mmol, 1 00 equiv) in dichloromethane (5 mL) was added tπethylamme (96 mg, 0 95 mmol, 2 99 equiv) and the solution cooled to O0C Intermediate 63 2 (407 mg, 0 96 mmol, 3 02 equiv) in dichloromethane (5 mL) was then added dropwise and the reaction was stirred for 1 h at room temperature The resulting mixture was concentrated under vacuum, the residue applied onto a silica gel column and then eluted with ethyl acetate/petroleum ether (1 2) to afford 150 mg (62%) of Intermediate 63 3 as a yellow solid
Figure imgf000245_0001
Intermediate 63.4: (2R,3R,4S,5R)-6-(4-(3-(6-chloro-2-(dianiinomethyleneamino)- quinazolin-4-yl)phenyl)piperazin-l-yl)-6-oxohexane-l,2,3,4,5-pentayl pentaacetate To Intermediate 63 3 (150 mg, 0 20 mmol, 1 00 equiv) in NMP (5 mL) was added guamdme (0 8 mL of a 1 moI/L solution in NMP, 40 equiv) and 1,4-diaza- bicycloβ 2 2]octane (DABCO, 44 8 mg, 040 mmol, 2 00 equiv) and the resulting solution was stirred for 1 5 h at 300C The reaction was quenched by the addition of 10 mL of water and then extracted with 2x10 mL of ethyl acetate The organic layers combined, dπed over anhydrous sodium sulfate, concentrated, applied onto a silica gel column and then eluted with dichloromethane/methanol (10 1) to afford 30 mg (19%) of Intermediate 63 4 as a yellow solid
Figure imgf000246_0001
Compound 63: 2-(6-chloro-4-(3-(4-((2R,3S,4R,5R)-2,3,4,5,6- pentahydroxyhexanoyl)piperazin-l-yl)phenyl)quinazolin-2-y])guanidine To
Intermediate 63 4 (25 mg, 0 03 mmol, 1 00 equiv) in methanol (5 mL), was added a solution of LiOH (3 9 mg, 0 16 mmol, 5 03 equiv) in water (02 mL) and the resulting solution was stirred for 0 5 h at O0C The pH value of the solution was adjusted to 7 with aqueous HCl (5 %), the resulting mixture was concentrated under vacuum and then purified by Prep-HPLC to afford 10 mg (45%) a TFA salt of compound 63 as a yellow solid LCMS (ES, m/z) 560 0 [M+H]+, 1H-NMR (300 MHz, CD3OD, ppm) 7 96-8 09 (m, 3H), 7 52-7 57 (m, IH), 7 25-7 39 (m, 3H), 4 73 (d, JS 1 Hz, IH), 4 07-4 09 (m, IH), 3 62-3 89 (m, 8H) MS (ES, m/z) 560 0 [M+H]+
Example 64
3-(4-(3-(6-chloro-2-(diamlnomethyleneamino)quinazolin-4-yl)pheny])piperazin-l- yl)propanoic acid
Figure imgf000246_0002
Intermediate 64.1: methyl 3-(4-(3-(2,6-dichloroquinazolin-4-yl)phenyl)piperazin- l-yl)propanoate To Intermediate 60 6 (200 mg, 0 51 mmol, 1 00 equiv) in tetrahydrofuran (10 mL) was added methyl acrylate (253 mg, 2 94 mmol, 5 81 equiv) and tnethylamme (253 mg, 2 50 mmol, 4 95 equiv) and the resulting mixture was stirred for 3 h at room temperature The reaction was concentrated under vacuum, the residue applied onto a silica gel column and then eluted with ethyl acetate/petroleum ether (1 3) to afford 100 mg (44%) of Intermediate 64.1 as a yellow solid
Figure imgf000247_0001
Compound 64 : 3-(4-(3-(6-chloro-2-(diaminomethyleneamino)quinazolin-4- yl)phenyl)piperazin-l-yl)propanoic acid. Compound 64 was prepared from Intermediate 64 1 usmg the procedures descnbed in Example 61, affording 25 mg of the title compound as a yellow solid , 1H-NMR (300 MHz, DMSO-d6, pprri) δ 7 89-7 92 (m, 3H), 7 42-7 47 (m, IH), 7 35(brs, IH), 7 15-7 24 (m, 2H), 3 25 (brs, 4H), 2 63-2 74 (m, 6H), 2 31-2 35 (m, 2H) LCMS (ES, m/z) 454 0 [M+H]+
Example 65 1 -(4-(3 -(4-(3-aminoρroρyl)piperazm- 1 -yl)phenyl)-6-chloroquinazolm-2-yl)guanidine
Figure imgf000247_0002
Compound 65: l-(4-(3-(4-(3-aminopropyl)piperazin-l-yl)phenyI)-6- chloroquinazolin-2-yl)guanidine A hydrochloride salt of the title compound was prepared using procedures similar to those outlined in Example 61, starting with intermediate 60 6 and tert-butyl 3-bromopropylcarbamate MS (ES, m/z) 439 [M+H]+ Example 66
4-(4-(6-chloro-2-(diaminomethyleneammo)quinazolin-4-yl)phenyl)piperazine-l- carboximidamide
Figure imgf000248_0001
Compound 66: 4-(4-(6-chloro-2-(dianiinomethyleneamino)quinazolin-4- yl)phenyl)piperazine-l-carboximidamide. A TFA salt of Compound 66 was prepared from Intermediate 61.1, using the procedures described in Example 60. MS (ES, m/z): 10 424 [M+H]+
Example 67
2-(4-(3-(4-(3-guanidinopropyl)piperazin-l-yl)phenyl)-6-chloroquinazolin-2- yl)guanidine
15
Figure imgf000248_0002
Compound 67: 2-(4-(3-(4-(3-guanidinopropyI)piperazin-l-yl)phenyl)-6- chloroquinazolin-2-yl)guanidine. A hydrochloride salt of Compound 67 was prepared from Compound 65 using the procedures outlined in Example 60. MS (ES, m/z): 481 [M+H]+
Example 68
2-(6-chloro-4-(3-(4-(2-hydroxyethyl)piperazin-l-yl)phenyl)quinazolin-2- yl)guanidine
Figure imgf000249_0001
Compound 68: 2-(6-chloro-4-(3-(4-(2-hydroxyethyl)piperazin-l- yl)phenyl)quinazolin-2-yl)guanidine. A TFA salt of Compound 68 was prepared from Compound 60.6 and ethylene oxide using the procedures outlined in Example 61. MS (ES, m/z): 426 [M+H]+
Example 69
2-(6-chloro-4-(4-(4-(2-hydroxyethyl)piperazin-l-yl)phenyl)quinazolin-2- yl)guanidine
Figure imgf000249_0002
Compound 69: 2-(6-chloro-4-(4-(4-(2-hydroxyethyl)piperazin-l- yl)phenyl)quinazolin-2-yl)guanidine a TFA salt of Compound 69 was prepared from Intermediate 61 1 using the procedures descπbed in Example 68 MS (ES, m/z) 426 [M+H]+
Example 70
4-(4-(3-(6-chloro-2-(diaminomethyleneamino)quinazolin-4-yl)phenyI)piperazin-l- yl)butanoic acid 2,2,2-trifluoroacetic acid salt
Figure imgf000250_0001
Compound 70: 4-(4-(3-(6-chloro-2-(diaminomethyleneamino)quinazolin-4- yl)phenyl)piperazin-l-yl)butanoic acid Compound 70 was prepared from Intermediate 60 6 and methyl 4-bromobutanoate using the procedures descπbed in Example 61 Purification by silica gel column with methanol water (0—0 04)gave a TFA salt of the title compound as a yellow solid 1H-NMR (300MHz, DMSO, ppm) δ 11 33(s, IH), 8 09-8 19(m, 2H), 7 96-7 96(s, IH), 7 53-7 58(m, IH), 7 25-7 37(m, 3H), 4 0(s, 4H), 3 16(s, 6H), 2 34-2 39(m, 2H), 1 92(s, 2H), MS (ES, m/z) 468 [M+H]
Examples 71-104
Examples 71 - 104 were prepared using methods descπbed in Examples 1-70 Charactenzation data (mass spectra) for compounds 71-104 are provided in Table 3
Example 71 (E)-3-(4-(4-(3-(diaminomethyleneamino)-2-methyl-3-oxoprop-l-enyl)-2,6- diflιiorophenoxy)phen\isιilfonaniido)pιopane- 1 -sulfonic acid
Figure imgf000251_0001
Example 72
2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinoIin-4-yl)-N- (phosphonomethyl)phenylsulfonamido)acetic acid
Figure imgf000251_0002
Example 73 4-(4-(4-(6-chloro-2-(diaminomethyleneamino)quinazolin-4-yl)phenyl)piperazin-l- yl)butanoic acid
Figure imgf000251_0003
Example 74 (E)-N-(diaininomethylene)-3-(4-(4-(N-(ethylcarbamoyl)sulfamoyl)phenoxy)-3,5- difluorophenyI)-2-methylacrylamide
Figure imgf000252_0001
Example 75
(E)-N-(diaminomethylene)-3-(4-(4-(N-(2-
(dimethylamino)ethylcarbamoyl)sulfamoyI)phenoxy)-3,5-difluorophenyl)-2- methylacrylamide
Figure imgf000252_0002
Example 76
4-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)phenylphosphonic acid
Figure imgf000252_0003
?.H0H
Example 77 (E)-N-(diamlnomethyIene)-3-(3,5-difluoro-4-(4-(N-methyl-N-((2S,3R,4R,5R)-,3,4,5,6-pentahydroxyhexyl)sulfamoyl)phenoxy)phenyl)-2-methylacrylaniide
Figure imgf000253_0001
Example 78
3-(4-(6,8-dichloro-2-methyI-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsu]fonamido)propane-l-sulfonic acid
Figure imgf000253_0002
Example 79
2-(4-(4-(4-(3-aminopropyl)piperazin-l-yl)phenyl)-6-chloroquinazolin-2- yl)guanidine
Figure imgf000253_0003
Example 80 3-(6,8-dichloro-2-methyI-l,2,3,4-tetrahydroisoquinolin-4-yl)-N-(2-(2-(2-(2-(4-
(hydroxymethyl)-lH-l,2,3-triazol-l- yl)ethoxy)ethoxy)ethoxy)ethyl)benzenesulfonamide
Figure imgf000254_0001
Example 81
N,N'-(2,2'-(2,2'-(2,2'-(2,21-(4,4'-oxybis(methylene)bis(lH-l,2,3-triazole-4,l- diyl))bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l- dlyl))bis(oxy)bis(ethane-2,l-diyl))bis(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide)
Figure imgf000254_0002
Example 82 N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide
Figure imgf000254_0003
Example 83 l-(2-(2-(2-(2-(3-(6,8-dichIoro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-lH-l,2,3-triazole-4,5- dicarboxylic acid
Figure imgf000255_0001
Example 84
(E)-3-(4-(4-(N-(2-(2-(2-aminoethoxy)ethoxy)ethyl)sulfamoyl)phenoxy)-3,5- difluorophenyl)-N-(diaminomethylene)-2-methylacrylamide
Figure imgf000255_0002
Example 85 2-(4-(4-(4-(2-aminoethyl)piperazin-l-yl)phenyl)-6-chloroquinazolin-2-yl)guanidine
Figure imgf000255_0003
Example 86 (E)-3-(4-(4-(N-(2-(2-(2-(2- ammoethoxy)ethoxy)ethoxy)ethylcarbamoyl)sulfamoyl)phenoxy)-3,5- difluorophenyl)-N-(diaminomethylene)-2-methylacrylamide
Figure imgf000256_0001
Example 87
Nl,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyH,2,3,4-tetrahydroisoquinolin-4- yl)phenylsuIfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide
Figure imgf000256_0002
Example 88
Nl,N4-bis(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide
Figure imgf000256_0003
Example 89 l-(4-(4-(4-(3-guanidinopropyl)piperazln-l-yl)phenyl)-6-chloroquinazolin-2- yl)guanidine
2S4
Figure imgf000257_0001
Example 90
(E)-2-(4-(2-(4-(4-(3-(diaminomethyleneamino)-2-methyl-3-oxoprop-l-enyl)-2,6- difluorophenoxy)phenylsulfonamido)ethyl)piperazin-l-yl)acetic acid
Figure imgf000257_0002
Example 91 N-(l-amino-l-imino-5,8,l l-trioxa-2-azatridecan-13-yl)-3-(6,8-dichloro-2-methyl- l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide
Figure imgf000257_0003
Example 92
N-(l-amino-l-imino-5,8,ll-trioxa-2-azatridecan-13-yl)-4-(6,8-dichloro-2-methyl- l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide
Figure imgf000258_0001
Example 93 (E)-l-(3-(3,5-difluoro-4-phenoxyphenyl)-2-methylalIyI)guanidine
DIBAL-H
Figure imgf000258_0002
Figure imgf000258_0003
Intermediate 93.1 (E)-3-(3,5-difluoro-4-phenoxyphenyl)-2-methylprop-2-en-l-ol:
To a solution of (E)-ethyl 3-(3,5-difluoro-4-phenoxyphenyl)-2-methylacrylate (Intermediate 41 2) (800mg, 2 51mmol) in dry DCM (25mL) under N2 at -78°C was added a solution of DIBAL-H (8 79mL, IM in DCM) dropwise over several minutes The reaction was allowed to warm to room temperature over 2 hours The reaction mixture was cooled to 00C, quenched with 25 mL of Rochelle's Salt solution (10% w/v in water), and stirred vigorously for 1 hour The resulting suspension was diluted with water (2OmL) and extracted with DCM (3x30mL) The combined organic layers were dπed over Na2SCv and concentrated The resulting oil was applied onto a silica gel column (50% EtOAc in hexanes) to yield 566mg of the title compound (82%) as a yellow oil
Figure imgf000258_0004
Intermediate 93.2 (E)-2-(3-(3,5-difluoro-4-phenoxyphenyl)-2- methylallyl)isoindoline-l,3-dione: To a solution of (E)-3-(3,5-difluoro-4- phenoxyphenyl)-2-methylprop-2-en-l-ol (Intermediate 93 1) (410mg, 1 49mmol) in dry toluene (7 45mL) under N2 was added PPI13 and phthahmide The resulting solution was cooled to O0C and diethyl azodicarboxylate (DEAD, 0 748mL) was added dropwise over several minutes The reaction was allowed to warm to room temperature and stirred overnight After diluting with EtOAc (2OmL), the organic layer was washed with water (2x30mL), bπne (3OmL) and dried over Nβ24 After removal of solvent, the resulting residue was applied to a silica gel column (15% EtOAc in hexanes) to yield 385mg of the title compound (63%) as an oil
Figure imgf000259_0001
Intermediate 93.3 (E)-3-(3,5-difluoro-4-phenoxyphenyl)-2-methylprop-2-en-l- amine: To a solution of (E)-2-(3-(3,5-difluoro-4-phenoxyphenyl)-2- methylallyl)isoindolme-l,3-dione (Intermediate 93 2, lOOmg, 025mmol) in methanol (ImL) was added hydrazine hydrate (25mg, 0 5mmol) and the reaction stirred at 500C overnight The white solid was filtered with DCM, and the solvent removed from the filtrate The residue was brought up in DCM and filtered This was repeated until no further precipitate formed to give 49 5mg of the title compound (71%) as a yellow oil, a lOmg portion of which was diluted with IN HCl and freeze dπed to give 7 8mg of the title compound as an HCl salt 1H-NMR (400MHz, d6-DMSO) δ 8 25 (s, 2H), 7 37 (t, 2H), 7 20 (d, 2H), 7 12 (t, IH), 6 97 (s, IH), 3 57 (s, 2H), 1 96 (s, 3H) MS (m/z) 258 96 (M-NH2)
Figure imgf000259_0002
Intermediate 93.4: (E)-4-(4-(3-amino-2-methylprop-l-enyl)-2,6-difluorophenoxy)- N-(2-(dimethylamino)ethyl)benzenesulfonamide: To a solution of (E)-3-(3,5- di£luoro-4-phenoxyphenyl)-2-methylρrop-2-en-l -amine (intermediate 93 3, lOOmg, 0 364mmol) in DCM (0 364 mL, IM) was added chlorosulfomc acid (2 91mmol, 194 3uL) m 4 portions dropwise every 20 minutes The reaction was stirred an additional 20 minutes and then quenched into a solution of Nl,Nl-dimethylethane-l,2- diamine (3 78mL) in DCM (12mL) at O0C The resulting solution was warmed to room temperature and stirred for 30 minutes Upon completion the solvent was removed and the residue brought up in 1 1 Acetonitπle water solution and purified by preparative HPLC to give 74 5mg of the title compound (31 %) as a TFA salt
Figure imgf000260_0001
Compound 93: (E)-4-(2,6-difluoro-4-(3-guanidino-2-methylprop-l-enyl)phenoxy)- N-(2-(dimethylamino)ethyl)benzenesulfonamide: To a solution of (E)-4-(4-(3- amino-2-methylprop- 1 -enyi)-2,6-difluorophenoxy)-N-(2-
(dimethylamino)ethyl)benzenesulfonamide (Intermediate 93 4, 39 3mg, 0 092mmol) m dry THF (46OuL, 02M) under N2 was added TEA (0 276HmIoI, 27 9mg) and (IH- pyrazol-l-yl)methanediamine hydrochlonde (0 102mmol, 14 9mg) The resulting solution was stirred for 1 hour, at which point LCMS indicated complete conversion The solvent was removed and the resulting residue brought up in 1 1 ACN water and purified by preparative HPLC to give 16 9mg of the title compound (26%) as a TFA salt 1H-NMR (400MHz, CD4OD) δ 7 87 (d, 2H), 7 12 (d, 2H), 7 08 (d, 2H), 3 92 (s, 2H), 3 62 (m, 2H), 3 29 (m, 2H), 3 17 (t, 2H), 2 01 (s, 6H), 1 91 (s, 3H) MS (m/z) 468 12 (M+H)+
Example 94 N-(2-(2-(2-(2-(4,5-bis(hydroxymethyl)-lH-l,2,3-triazol-l- yl)ethoxy)ethoxy)ethoxy)ethyI)-3-(6,8-dichloro-2-methyI-l,2,3,4- tetrahydroisoquinoIin-4-yl)benzenesulfonamide
Figure imgf000261_0001
Example 95
N-(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)acetamide
Figure imgf000261_0002
Example 96
N-(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)acetamide
Figure imgf000261_0003
Example 97 Nl,N31-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-4,7,10,13,16,19,22,25,28- nonaoxahentriacontane-l,31-diamide °
Figure imgf000262_0001
Example 98
Nl,N31-bis(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-4,7,10,13,16,19,22,25,28- nonaoxahentriacontane-l,31-diamide
Figure imgf000262_0002
IQ
Example 99
(E)-3-(4-(4-(N-(l-amino-l-imino-5,8,ll-trioxa-2-azatridecan-13- yl)sulfamoyl)phenoxy)-3,5-difluorophenyl)-N-(diaminomethylene)-2- methylacrylamide
Figure imgf000262_0003
Example 100
N,N'-(13-oxo-3,6,9,17,20,23-hexaoxa-12,14-diazapentacosane-l,25-diyl)bis(3-(6,8- 2G dichloro-2-methyl-l,2,3,4-tetrahydroisoquinoIin-4-yl)benzenesuIfonamide)
Figure imgf000262_0004
Example 101
(E)-N-(diaminomethylene)-3-(3,5-difluoro-4-(4-(N-(2-oxo-6,9,12-trioxa-3-
25 azatetradecan-14-yl)sulfamoyl)phenoxy)phenyl)-2-methylacrylamide
Figure imgf000263_0001
Example 102 Nl,N31-bis(2-(2-(2-(2-(4-(4-((E)-3-(diaminomethyleneamino)-2-methyl-3-oxoprop- l-enyl)-2,6-difluorophenoxy)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)- 4,7,10,13,16,19,22,25,28-nonaoxahentriacontane-l,31-diamide
Example 103
N,N'-(13-oxo-3,6,9,17,20,23-hexaoxa-12,14-diazapentacosane-l,25-diyl)bis(4-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide)
Figure imgf000263_0003
Example 104 Nl,N4-bis(20-(4-(4-((E)-3-(dlaminomethyleneamino)-2-methyl-3-oxoprop-l-enyl)-
2,6-difluorophenoxy)phenylsulfonamido)-3,6,9,12,15,18-hexaoxaicosyl)-2,3- dihydroxysuccinamide
Figure imgf000263_0004
Table 3
Figure imgf000264_0001
Table 3
Analytical Data for Example Compounds 71-104
Example [M+H]+
99 628
100 1117
101 628
102 1649
103 1117
104 1549
Example 105
4-/3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)-polyethylimino- sulfonamide
Figure imgf000265_0001
Example 105 is prepared from polyethylamme according to the procedures in descπbed in Examples 1-70, where "x," "y," "n" and "m" are determined by the stoichiometry of the sulfonyl chloride and polyethylamme
Example 106
As illustrated below, other polymeric nucleopmles are employed using the procedures descπbed m Examples 1-70 to prepare polyvalent compounds Other polymeric nucleophiles
Figure imgf000266_0001
Example 107
As illustrated below, polymeric electrophiles are used with nucleophilic Intermediates 5 to prepare polyvalent compounds using, for example, the procedures outlined in Example 68
Figure imgf000266_0002
10 Example 108-147
General Procedure for copolymerization of Intermediate 108.1 and Intermediate 108.2 with other monomers
Figure imgf000266_0003
I5
Intermediate 108 1 N-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolm-4- yl)phenyl)acrylamide Intermediate 108 1 (Int 108 1) was prepared from intermediate 307 and acrylic acid using procedures descπbed in Examples 1 - 70 MS (m/z) 361 1 (M+H) Intermediate 108 2 N-(2-(3-(6,8-dichloro-2-methyM,2,3,4-tetrahydroisoquinolm-4- yl)phenylamino)ethyl)acrylamide Intermediate 108 2 (Int 108 2) was prepared from intermediate 307 using procedures described in Examples 1 - 70 MS (m/z) 404 1 5 (M+H)
A 20-mL vial is charged with a total of Ig of Intermediate 108 1 or Intermediate 108 2 and other monomers, a total of 9g of isopropanol/dimethylformamide solvent mixture, and 20 mg of azobisisobutyromtnle The mixture is degassed for 1 mm and is sealed
10 under a nitrogen atmosphere The stoichiometry for each example is shown in Tablel The reaction mixture is heated in an oil bath to 700C under stirring After 8 h at 7O0C the reaction mixture is cooled down to ambient temperature and then 10 mL of water is added The solution is then transferred to a dialysis bag (MWCO 1000) for dialysis against DI water for 2 days The resulting solution is freeze-dπed to afford copolymers
I5
Figure imgf000267_0001
Figure imgf000268_0001
Figure imgf000269_0002
Example 148 Synthesis of 2-Methyl-acrylic acid 3-trimethyIsilany]-prop-2-ynyl ester
Figure imgf000269_0001
A solution of tnmethylsilyl propyn-1-ol (1 g, 7 8 mmol) and Et3N (1 4 mL, 10 mmol) in Et2O (10 mL) is cooled to -20 0C and a solution of methacryloyl chloride (0 9 mL, 9 3 mmol) in Et2O (5 mL) is added dropwise over 1 h The mixture is stirred at this temperature for 30 mm, and then allowed to warm to ambient temperature overnight Any precipitated ammonium salts can be removed by filtration, and volatile components can be removed under reduced pressure The crude product is then purified by flash chromatography
Examples 149-154
General Procedure for synthesis of poly N-(2-hydroxypropyl)methacrylamide-co- prop-2-ynyl methacrylate
Figure imgf000270_0001
General procedure for copolymeπzation of N-(2-hydroxypropyl)methacrylamide and 3- (tnmethylsilyl)prop-2-ynyl methacrylate
A 100-mL round bottom flask equipped with a reflux condenser is charged with a total 5g of N-(2-hydroxypropyl)methacrylamide and 3-(tπmethylsilyl)prop-2-ynyl methacrylate, 45g of lsopropanol/dimethylformamide solvent mixture, and 100 mg of azobisisobutyromtnle The mixture is degassed for 1 mm and maintained under nitrogen atmosphere during the reaction Stoichiometry for each example is shown in Table 5 The reaction mixture is heated in an oil bath to 70 0C under stirring, and after 8 h the reaction mixture is cooled to ambient temperature and then 30 mL of solvent is evaporated under vacuum The resulting solution is then precipitated into 250 mL of Et2θ The precipitate is collected, redissolved in 10 mL of DMF, and precipitated again into 250 mL of Et2O The resulting precipitate is dned under vacuum to afford copolymers
General procedure for removal of tπmethyl silyl group
The tnmethyl silyl protected polymer (4g), acetic acid (1 5 equiv mol/mol with respect to the alkyne-tπmethylsilyl groups), and 200 mL of THF is mixed in a 500 mL flask The mixture is cooled to -20 0C under nitrogen atmosphere and followed by addition of 0 20 M solution of tetra-n-butylammonium fluoride tπhydrate (TBAF 3H2O) in THF (1 5 equiv mol/mol with respect to the alkyne-tπmethylsilyl groups) over a course of 5 mm The solution is stirred at this temperature for 30 mm and then warmed to ambient temperature for an additional 8 hours The resulting mixture is passed through a short silica pad and then precipitated in Et2θ The resulting precipitate is dπed under vacuum to afford copolymers
Figure imgf000271_0001
Examples 154-167
General procedure for post-modification of Examples 149-153 by [2+3] cycloaddition
Figure imgf000272_0001
Polymer 154 (54 mg) containing 0 1 mmol of alkyne moiety, a total of 0 1 mmol of azido-compounds (Intermediate 28 1, 13-azido-2,5,8,l l-tetraoxatndecane, N (2- azidoethyl)-3-(dimethylamino)propanamide and 1-azidodecane, corresponding ratios shown in Table 6), 0 05 mmol of dnsopropylethylamine, and 1 mL of DMF is mixed at ambient temperature and degassed for 1 mm While maintaining a nitrogen atmosphere, copper iodide (10 mg, 0 01 mmol) is then added to the mixture The solution is stirred at ambient temperature for 3 days and then passed through a short neutral alumina pad The resulting solution is diluted with 10 mL of DI water, dialyzed against DI water for 2 days, and lyophihzed to afford copolymers
Figure imgf000272_0002
Figure imgf000273_0002
Example 168
Nl,N4-bis(2-(2-(2-(3-(6,8-dichloro-2-methyI-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethyI)-2,3-dihydroxysuccinaniide
Figure imgf000273_0001
Intermediate 168.1, bis(2,5-dioxopyrrolidin-l-yl) 2,3-dihydroxysuccinate To a
500 ml 3-necked roundbottom flask was added 2,3-dihydroxysuccimc acid (10 0 g, 66 62 mmol, 1 00 eqmv), N,N'-Dicyclohexyl carbodπmide (DCC, 300 g, 145 42 mmol, 2 18 equiv) and tetrahydrofuran (THF, 100 mL) This was followed by the addition of a solution of N-hydroxysuccinimide (NHS, 16 5 g, 143 35 mmol, 2 15 equiv) m THF (100 mL) at 0-100C The resulting solution was warmed to room temperature and stirred for 16 h The solids were filtered out and the filtrate was concentrated under vacuum The crude product was re crystallized from N,N- dimethylfbrmamide (DMF) / ethanol in the ratio of 1 10 This resulted in 5 2 g (22%) of the title compound as a white solid 1H-NMR (300MHz, DMSO, ppm) δ 6 70(d, J=I 8Hz, 2H), 4 89(d, J=I 2Hz, 2H), 2 89(s, 8H) MS (w/z) 367 [M+Na]+
Figure imgf000274_0001
Intermediate 168.2 N-(2-(2-(2-aminoethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide: To a 50-mL 3- necked round-bottom flask was added 2-(2-(2-ammoethoxy)ethoxy)ethanamme (3 2 g, 21 59 mmol, 21 09 eqmv) and dichloromethane (DCM, 20 mL) This was followed by the addition of a solution of 3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolm-4- yl)benzene-l-sulfonyl chlonde (Intermediate 1 6) (400 mg, 1 02 mmol, 1 00 equiv) in DMF (5 mL) dropwise with stirring The resulting solution was stirred for 5 h at which time it was diluted with 100 mL of ethyl acetate The resulting mixture was washed successively with 2x10 mL of water and 1x10 mL of Bnne The organic layer was dπed over anhydrous sodium sulfate and concentrated under vacuum This resulted in 300 mg (58%) of N-(2-(2-(2-ammoethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolm-4-yl)benzenesulfonamide as a yellow oil
Figure imgf000274_0002
Compound 168, Nl,N4-bis(2-(2-(2-(3-(6,8-dichloro-2-methyM,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide: Into a 50-mL round-bottom flask was placed a solution of N- (2-(2-(2-ammoethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide (300 mg, 0 60 mmol, 1 00 equiv) in DMF (5 ml), bis(2,5-dioxopyrrolidm-l-yl) 2,3-dihydroxysuccinate (92 5 mg, 027 mmol, 045 equiv) and tnethylamme (TEA, 1 0 g, 9 88 mmol, 16 55 equiv) The resulting solution was stirred overnight at room temperature and then concentrated under vacuum The crude product (300 mg) was purified by Prep-HPLC with the following conditions Column, SunFire Prep C18, 5um, 19*150mm, mobile phase, Water with 0 05%TFA and CH3CN (20% CH3CN up to 40% in 5 mm, up to 100% in 2 mm), Detector, uv 220&254nm This resulted in 192 4 mg (28%) of a TFA salt of the
10 title compound as a white solid 1H-NMR (300MHz, DMSO, ppm) δ 7 92 (d, J=I 8Hz, 2H) , 7 82 (m, 2H) , 7 67 (t, J=I 8Hz, 2H) , 7 57 (m, 2H) , 7 55 (d, J-6 9Hz, 2H) , 6 86 (m, 2H) , 4 84(s, 2H), 4 79(s, 2H) , 4 54(d, 2H), 448(s, 2H), 3 92(m, 2H) , 3 53(m, 22H) , 3 18(s, 6H), 3 07(t, /-5 4Hz, 4H) MS (m/z) 1119 [M+H]+
I5
Example 169
Nl,N4-bis(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethyl)-2,3-dihydroxysuccinamide
Figure imgf000275_0001
Intermediate 169.1, N-(2-aminoethyl)-3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide: Into a 50-mL 3-necked round- bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a 25 solution of 3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinohn-4-yl)benzene-l- sulfonyl chlonde (intermediate 1 6) (100 mg, 0 26 mmol, 1 00 equiv) in DCM (5 mL) This was followed by the addition of a solution of ethane 1,2-diamme (307 mg, 5 11 mmol, 19 96 equiv) in DCM / DMF (10/1 mL) The resulting solution was stirred for 5 h at room temperature The mixture was concentrated under vacuum The resulting solution was diluted with 50 mL of ethyl acetate and washed with 2x10 mL of water and then 1x10 mL of Bπne The organic layer was dried over anhydrous sodium sulfate and concentrated under vacuum to afford 90 mg (76%) of N-(2-aminoethyl)-3-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolm-4-yl)benzenesulfonamide as yellow
Figure imgf000276_0001
Compound 169, Nl,N4-bis(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroiso- quinolin-4-yl)phenyIsu]fonamido)ethyl)-2,3-dihydroxysuccinamide: Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of N-(2-ammoethyl)-3-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoquinohn-4-yl)benzenesulfonamide (250 mg, 0 60 mmol, 1 00 equiv) in DMF (5 mL), bis(2,5-dioxopyrrolidm-l-yl) 2,3-dihydroxysuccinate (Intermediate 168 1) (92 mg, 0 27 mmol, 044 equiv) and tπethylamme (280 mg, 2 77 mmol, 4 55 equiv) and the resulting solution was stirred overnight at room temperature The resulting mixture was concentrated under vacuum, the residue diluted with 100 mL of ethyl acetate and then washed with 2x10 mL of water The organic layer was dned over anhydrous sodium sulfate and concentrated under vacuum The crude product was purified by Prep-HPLC with the following conditions Column, SunFire Prep Cl 8, 5um, 19* 150mm, mobile phase, Water with 0 05%TFA and CH3CN (25% CH3CN up to 35% in 5 mm, up to 100% in 2 5 mm), Detector, uv 220&254nm This resulted in 88 4 mg (15%) of a TFA salt of Nl,N4-bis(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoqumolin-4-yl)phenylsulfonamido)ethyl)-2,3-dihydroxysuccinamide as a white solid 1H-NMR (400MHz, CD3OD, ppm) § 1 61 (d, J=I 6Hz, 2H), 7 61(s, 2H), 744 (t, J=I 6Hz, 2H) , 737 (d, J=I 6Hz, 2H), 7 25 (d, J=2Hz, 2H), 6 72 (s, 2H) , 4 33 (t, J=64Hz, 2H) , 4 30 (s, 2H), 3 64 (m, 4H), 3 21(s, 4H) , 2 98 (m, 2H), 2 90(m, 4H), 2 65 (m, 2H) , 2 42 (s, 6H) MS (ικ/z) 943 [M+H]+
Example 170
Nl^-bistZ-Ca^Z-tZ-CS^ό^-dichloro-l-ethyl-l^^^-tetrahydroisoquinolin^- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysucclnamide
Figure imgf000277_0001
Intermediate 170.1, 3-(6,8-dichloro-2-ethyl-l,2,3,4-tetrahydroisoquinolin-4- yl)benzene-l-sulfonyl chloride: Using procedures outlined m Example 1 to prepare intermediate 1 6, substituting N-(2,4-dichlorobenzyl)ethanamine for l-(2,4- dichlorophenyl)-N-methylmethanamine, the title compound was prepared as a hydrochloπde salt
Figure imgf000277_0002
Intermediate 170.2 N-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8- dichloro-2-ethyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesuIfonamide To 2-(2- (2-(2-azidoethoxy)ethoxy)ethoxy)ethanamine (300 mg, 1 51 mmol, 1 00 equiv) in DCM (10 mL) was added TEA (375 mg, 3 00 equiv) followed by the portionwise addition of 3-(6,8-dichloro-2-ethyl-l,2,3,4-tetrahydroisoqumolm-4-yl)benzene-l-sulfonyl chloride (500 mg, 1 23 mmol, 1 00 equiv) The resulting solution was stirred for 1 h at room temperature and then concentrated under vacuum The residue was applied onto a silica gel column and eluted with ethyl acetate/petroleum ether (1 2) to afford 04 g (41%) of N-(2-(2-(2 (2~azidoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-ethyl-l,2,3,4- tetrahydroisoqumolm-4-yl)benzenesulfonamide as yellow oil
Figure imgf000278_0001
Intermediate 170.3, N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8- dichloro-2-ethyl-l,2,3,4-tetrahydroisoquinolin-4-yl)beπzenesulfoπamide: Into a 100-mL round-bottom flask, was placed N-(2-(2-(2-(2- azidoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-ethyl-l, 2,3,4- tetrahydroisoqumolin-4-yl)benzenesulfonamide (400 mg, 0 68 mmol, 1 00 equiv), tπphenylphosphme (400 mg, 2 20 equiv), THF (10 mL) and water(l mL) and the reaction was stirred overnight at room temperature The resulting mixture was concentrated under vacuum and applied onto a preparative thm-layer chromatography (TLC) plate, elutmg with DCM methanol(5 1) This resulted in 350 mg (73%) of N-(2- (2-(2-(2-ammoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-ethyl-l,2,3,4- tetrahydroisoquinolm-4-yl)benzenesulfonamide as yellow oil
Figure imgf000279_0001
Compound 170, Nl,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-ethy]-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide: Into a 50-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of N-(2-(2-(2- (2-aminoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-ethyl-l,233,4- tetrahydroisoqumolm-4-yl)benzenesulfbnarmde (100 mg, 0 18 mmol, 1 00 equiv) in DMF(3 mL), bis(2,5-dioxopyrrolidm-l-yl) 2,3-dihydroxysuccmate (Intermediate 168 1) (25 mg, 0 07 mmol, 045 equiv) and tnethylamine (75 mg, 4 50 equiv) The resulting solution was stirred overnight at room temperature The reaction progress was monitored by LCMS The resulting mixture was concentrated under vacuum The crude product was purified by Flash-Prep-HPLC with water methanol (1 10-1 100) This resulted in 12 1 mg (5%) of Nl,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-ethyt-l,2,3,4- tetrahydroisoqumolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3 - dihydroxysuccinamide as yellow oil 1H-NMR (300MHz, DMSO, ppm) δ 7 70- 7 60(m, 8H), 7 53-7 49(m, 6H), 6 88(s, 2H), 5 61-5 59(m, 2H), 4 38(m, 2H), 424- 4 22(m, 2H), 3 78-3 72(m, 2H), 3 58-3 48(m, 2H), 3 43(m, 7H), 3 43-3 40(m, HH), 3 27-3 20(m, 5H), 2 91-2 87(m, 6H), 2 76-2 70(m, 2H), 2 61-2 55(m, 3H), 1 04-0 99(m, 6H) MS (m/z) 1235 [M+H]+
Example 171
3,3'-(2,2'-(2,2'-(2,2'-oxybis(ethane-2,l-diyl)bis(oxy))bis(ethane-2,l-diyI))bis(6,8- dichloro-l,2,3,4-tetrahydroisoquinoline-4,2-diyl))dianiline
Figure imgf000280_0001
Intermediate 171.1, 2-(2,4-dichlorobenzylamino)-l-(3-nitrophenyl)ethanone: Into a 250-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 2-bromo-l-(3-mtrophenyl)ethanone (10 0 g, 41 15 mmol, 1 00 eqmv) in THF (150 mL), (2,4-dichlorophenyl)methanamme (7 16 g, 40 91 mmol, 1 00 equiv) and tnethylamme (5 96 g, 59 01 mmol, 1 50 eqmv) The resulting solution was stirred for 2 h at 250C The solids were filtered out The filtrate was concentrated to dryness and used for next step, assuming theoretical yield
Figure imgf000280_0002
Intermediate 171.2, 2-(2,4-dichlorobenzyIamino)-l-(3-nitrophenyl)ethanol: Into a 500-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of intermediate 171 1 (4091 mmol, 1 00 eqmv) in methanol (150 mL) This was followed by the addition Of NaBH4 (2 5 g, 65 79 mmol, 1 50 equiv) in several batches at O0C The resulting solution was stirred for 2 h at 250C The reaction was then quenched by the addition of aqueous NH4Cl The resulting mixture was concentrated under vacuum, and the solids were collected by filtration The crude product was purified by re-crystalhzation from ethyl acetate This resulted in 3 5 g (23%) of 2 (2,4-dichlorobenzylamino)-l-(3-mtrophenyl)ethanol as a yellowish solid
Figure imgf000280_0003
Intermediate 171.3, 6,8-dichloro-4-(3-nitrophenyl)-l,2,3,4-tetrahydroisoquinoline:
To 2-(2,4-dichlorobenzylamino)-l-(3-mtrophenyl)ethanol (intermediate 171 2) (500 mg, 1 47 mmol, 1 00 equiv) in DCM (10 mL) was added cone sulfuric acid (4 mL) dropwise with stirring at 0-50C The resulting solution was stirred for 12 h at room temperature The reaction was then quenched by the addition of water/ice The pH value of the solution was adjusted to 10 with sodium hydroxide The resulting solution was extracted with 2x50 mL of DCM and the organic layers combined and dπed over anhydrous sodium sulfate and concentrated under vacuum This resulted m 300 mg (63%) of 6,8-dichloro-4-(3-mtrophenyl)-l,2,3,4-tetrahydroisoquinohne as yellow oil
TsCI
TsO OTs DCM
Intermediate 171.4, 2,2'-(2,2'-oxybis(ethane-2,l-diyl)bis(oxy))bis(ethane-2,l-diyI) bis(4-methylbenzenesulfonate): Into a 250-mL 3-necked round-bottom flask, was placed a solution of tetraethylene glycol (10 g, 51 55 mmol, 1 00 equiv) in DCM (100 mL) This was followed by the addition of a solution of 4-methylbenzene-l-sulfonyl chloπde (21 4 g, 112 63 mmol, 2 20 equiv) in DCM (50 mL) dropwise with stirring at 5°C To this was added N,N-dimethylpyπdin-4-amine (15 7 g, 128 69 mmol, 2 50 equiv) The resulting solution was stirred for 2 h at room temperature at which time it was diluted with 100 mL of water The resulting solution was extracted with 3x100 mL of DCM and the organic layers combined The resulting mixture was washed with 1x100 mL of brine and then concentrated under vacuum The residue was applied onto a silica gel column and eluted with ethyl acetate/petroleum ether (1 2) to afford H g (43%) of the title compound as white oil
Figure imgf000281_0001
Intermediate 171.5, 2,2'-(2,2'-(2,2'-oxybis(ethane-2,l-diyl)bis(oxy))bis(ethane-2,l- diyl))bis(6,8-dichloro-4-(3-nitrophenyl)-l,2,3,4-tetrahydroisoquinoline: To 6,8- dichloro-4-(3-mtrophenyl)-l,2,3,4-tetrahydroisoqumoline (intermediate 171 3) (171 mg, 0 53 mmol, 2 50 eqmv) in DMF (2 mL) was added potassium carbonate (87 mg, 5 063 mmol, 3 00 equiv) and intermediate 171 4 (106 mg, 021 mmol, 1 00 equiv) and the resulting solution was stirred at 5O0C After stirring overnight, the resulting solution was diluted with 20 ml of water The resulting mixture was extracted with 3x20 ml of ethyl acetate and the organic layers combined and concentrated under vacuum The crude product was purified by Prep-HPLC with methanol water (1 1) This resulted m 10 10 mg (2%) of 2,2'-(2,2'-(2,2'-oxybis(ethane-2,l-diyl)bis(oxy))bis(ethane-2,l- diyl))bis(6,8-dichloro 4-(3-mtrophenyl)-l,2,3,4-tetrahydroisoquinolme) as a light yellow solid
Figure imgf000282_0001
I5
Compound 171, 3,3'-(2,2'-(2,2'-(2,2'-oxybis(ethane-2,l-diyI)bis(oxy))bis(ethane-2,l- diyl))bis(6,8-dichloro-l,2,3,4-tetrahydroisoquinoline-4,2-diyl))dianiline: To intermediate 171 5 (50 mg, 0 06 mmol, 1 00 equiv) in ethanol (5 mL) was added iron (34 mg, 0 61 mmol, 9 76 equiv) followed by the addition of hydrogen chloπde (5 mL)
20 dropwise with stimng The resulting solution was stirred for 2 h at room temperature and then for an additional 4 h at 550C The reaction progress was monitored by LCMS The solids were filtered out and the resulting solution was diluted with 10 mL of water The resulting mixture was concentrated under vacuum and the pH of the solution was adjusted to 9-10 with sodium carbonate The resulting solution was extracted with 3x50
25 mL of ethyl acetate and the organic layers combined, washed with 50 mL of bnne and then concentrated under vacuum The crude product was purified by Prep HPLC with H2O CH3CN (10 1) This resulted in 5 mg (11%) of 3,3'-(2,2'-(2,2' (2,2' oxybis(ethane- 2)l-diyl)bis(oxy))bis(ethane-2,l-diyl))bis(6,8-dichloro-l ,2,3,4-tetrahydroisoquinolme- 4,2-diyl))dianilme as a yellow solid ) 1H-NMR (400MHz, CD3OD, ppm) δ 7 27 (m, 2H), 7 06 (m, 2H), 6 80 (s, 2H), 6 63 (d, 2H), 6 54 (m, 4H), 4 14 (m, 2H), 402 (d, 2H), 3 65(m, 12H), 3 19 (m, 3H), 2 81(s, 4H), 2 71 (m, 2H) MS (m/z) 745 [M+H]+
Example 172
Nl,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsuIfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide
Figure imgf000283_0001
0
Intermediate 28.1 : N-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro- 2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide: To 2-(2-(2-(2- azidoethoxy)ethoxy)ethoxy)ethanartlme (1 5 g, 6 87 mmol, 1 79 equiv) in DCM (20 mL) was added tπethylamme (1 5 g, 14 82 mmol, 3 86 equiv) and 3-(6,8-dichloro-2-5 methyl-l,2,3,4-tetrahydroisoqumohn-4-yl)benzene-l-sulfonyI chloπde (1 5 g, 3 84 mmol, 1 00 equiv) The reaction was stirred overnight at room temperature at which time the resulting mixture was concentrated under vacuum The residue was dissolved m 100 mL of ethyl acetate and then was washed with 2x20 mL of water, dried over anhydrous sodium sulfate and concentrated under vacuum This resulted in 1 8 g (85%)Q of N-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoqumohn-4-yl)benzenesulfonamide as yellow oil
Figure imgf000283_0002
Compound 28, N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethy])-3-(6,8-dichloro-2- methyI-l,2,3,4-tetrahydroisoquinolln-4-yl)benzenesulfonamide: To N-(2-(2-(2-(2- azidoemoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoqumolm-4-yl)benzenesulfonamide (1 8 g, 3 26 mmol, 1 00 equiv) in THF 5 (30 tiiL) was added tπphenylphosphme (2 6 g, 9 91 mmol, 3 04 equiv) The resulting solution was stirred overnight at room temperature and then concentrated under vacuum The crude product (5 0 g) was purified by Flash-Prep-HPLC with the following conditions Column, silica gel, mobile phase, methanol water=l 9 increasing to methanol water~9 1 within 30 min, Detector, UV 254 nm 1 2 g product was 10 obtained This resulted m 1 2 g (64%) of N-(2-(2-(2-(2- ammoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoqumohn-4-yl)benzenesulfonamide as yellow oil
Figure imgf000284_0001
15
Compound 172, NlJV4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinoHn-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide: To N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoquinohn-4-yl)benzenesulfonamide (compound
20 28) (1 2 g, 2 28 mmol, 1 00 equiv) in DMF (8 mL) was added bis(2,5-dioxopyrrohdin- 1-yl) 2,3-dihydroxysuccmate (intermediate 168 1) (393 mg, 1 14 mmol, 0 50 equiv) and tπethylamme (1 5 g, 14 82 mmol, 6 50 equiv) and the resulting solution was stirred overnight at room temperature The mixture was concentrated under vacuum and the crude product was purified by Flash-Prep-HPLC with the following conditions
25 Column, silica gel, mobile phase, methanol water~l 9 increasing to methanol water=9 1 within 30 mm, Detector, UV 254 nm This resulted in 591 mg (43%) of Nl,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolm-4-yl)phenylsulfonamido)emoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide as a light yellow solid 1H-NMR (300MHz, CD3OD, ppm) δ 5 7 92 (d, J=I 8Hz, 2H), 7 81 (m, 2H), 7 67 (t, J-I 8Hz, 2H , 7 57 (m, 2H), 7 55 (d, J=6 9Hz, 2H), 6 85 (m, 2H), 4 78 (s, 2H), 4 77 (s, 2H), 4 54(d, J=402Hz, 2H), 4 48(s, 2H), 3 92(m, 2H), 3 53(m, 30H), 3 18(s, 6H), 3 07 (t, J-S 4Hz, 4H) MS (m/z) 603 [1/2M+H]+
10 Example 173
Nl,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylamino)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide
Figure imgf000285_0001
I5
Intermediate 173.1, N-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoquinoHn-4-yl)aniline: Into a 10-mL round- bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 4 (3 bromophenyl) 6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinohne
20 (intermediate 1 4) (400 mg, 1 08 mmol, 1 00 equiv) in DMSO (6 mL), 2-(2-(2-(2- azidoethoxy)ethoxy)ethoxy)ethanamme (236 11 mg, 1 08 mmol, 1 00 equiv), (S)- pyrrolidine-2-carboxylic acid (24 79 mg, 0 21 mmol, 0 20 equiv), copper(I) iodide (2048 mg, 0 11 mmol, 0 10 equiv) and potassium carbonate (223 18 mg, 1 62 mmol, 1 50 equiv) The resulting solution was stirred at 9O0C m an oil bath and the reaction
25 progress was monitored by LCMS After stirring overnight the reaction mixture was cooled with a water/ice bath and then diluted with ice water The resulting solution was extracted with 3x30 mL of ethyl acetate and the organic extracts were combined and washed with 2x20 mL of bπne The mixture was dπed over anhydrous sodium sulfate and concentrated under vacuum The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (2 1) This resulted in 130 mg (24%) of N-(2-(2-(2-(2- azidoethoxy)emoxy)emoxy)ethyl)-3-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoquinolm-4-yl)benzenamine as yellow oil
Figure imgf000286_0001
Intermediate 173.2, N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethy])-3-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)aniline: Into a 50-mL round- bottom flask, was placed a solution of intermediate 173 1 (350 mg, 069 mmol, 1 00 equiv) in THF/water (4/0 4 niL) and tπphenylphosphme (205 mg, 0 78 mmol, 1 20 equiv) The resulting solution was stirred overnight at 400C in an oil bath The resulting mixture was then concentrated under vacuum The pH of the solution was adjusted to 2- 3 with IN hydrogen chloπde (10 ml) The resulting solution was extracted with 2x10 mL of ethyl acetate and the aqueous layers combined The pH value of the solution was adjusted to 11 with NH3 H2O The resulting solution was extracted with 3x30 mL of DCM and the organic layers combined The resulting mixture was washed with 30 mL of bπne The mixture was dned over anhydrous sodium sulfate and concentrated under vacuum This resulted m 250 mg (75%) of N (2-(2-(2-(2- aminoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)anilme as yellow oil
Figure imgf000287_0001
Compound 173, Nl,N4-bis(2-(2-(2-(2-(3-(6,8-dichIoro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylamino)ethoxy)ethoxy)ethoxy)ethyl)-2,3- 5 dihydroxysuccinamide: To intermediate 173 2 (240 mg, 0 50 mmol, 1 00 eqiuv) in DMF (5 niL) was added TEA (233 mg, 2 31 mmol, 4 50 equiv) and bis(2,5- dioxopyrrolidin-1-yl) 2,3-dihydroxybutanedioate (intermediate 168 1) (62 mg, 0 18 mmol, 0 35 equiv) and the resulting solution was stirred overnight at room temperature The resulting mixture was concentrated under vacuum and the crude product was
10 purified by Prep-HPLC with methanol water (1 10) This resulted in 140 mg (26%) of Nl,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinohn-4- yl)phenylamino)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamideas a white solid 1H-NMR (300MHz, DMSO, ppm) δ 7 65 (m, 4H), 7 11 (m, 2H), 6 83 (m, 2H), 6 58 (m, 2H), 641 (m, 4H), 4 09 (m, 32H), 3 45 (m, 17H), 3 43 (m, 5H), 3 31 (m, 9H),
15 2 51 (m, 6H) MS (Wz) 1079 [M+H]+
Example 174
Nl,N4-bis(l-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinoIin-4- yl)phenylamino)-l-oxo-5,8,ll-trioxa-2-azatridecan-13-yl)-2,3-
20 dihydroxysuccinamide
Figure imgf000288_0001
Intermediate 174.1, l-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-3-(3-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yI)phenyI)urea: To 4- mtrophenyl 3-(6,8-dichloro-2-methyl- 1 ,2,3 ,4-tetrahydroisoqumolin-4- yl)phenyl carbamate (prepared by the procedure descπbed in example 38) (200 mg, 040 mmol, 1 00 equiv, 95%) in DMF (5 mL) was added TEA (170 mg, 1 60 mmol, 4 00 equiv, 95%) and 2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethanamine (90 mg, 0 39 mmol, 1 00 equiv, 95%) and the resulting solution was stirred for 2 h The mixture was then concentrated under vacuum, diluted with 10 mL of water and then extracted with 3x30 mL of ethyl acetate The organic layers were combined, washed with 3x30 mL of bπne, dπed over anhydrous sodium sulfate and then evaporated The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 5~1 1) This resulted in 160 mg (72%) of l-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-3-(3-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoquinohn-4-yl)phenyl)urea as pale-yellow oil
Figure imgf000288_0002
Intermediate 174.2 l-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-3-(3-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)phenyl)urea: Intermediate 1742 was prepared from l-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-3-(3-(6,8- dichloro-2 methyl-1, 2,3,4 tetrahydroisoqumohn-4-yl)phenyl)urea (intermediate 174 1) using the procedure descπbed to prepare intermediate 173 2 The crude product was purified by silica gel chromatography, elutmg with DCM/methanol (50 1 ) This resulted m 230 mg of l-(2-(2-(2-(2-ammoethoxy)ethoxy)ethoxy)ethyl)-3-(3-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoqmnolm-4-yl)phenyl)urea as pale-yellow oil
Figure imgf000289_0001
Compound 174, Nl,N4-bis(l-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylamino)-l-oxo-5,8,ll-trioxa-2-azatridecan-13-yl)- 2,3-dihydroxysuccinamide: Compound 174 was prepared from l-(2-(2-(2-(2- ammoethoxy)ethoxy)ethoxy)ethyl)-3-(3-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoquinolm-4-yl)phenyl)urea (intermediate 1742) using the procedures descπbed m example 172 The crude product (400 mg) was purified by Prep-HPLC with methanol acetomtπle - 60 40 This resulted in 113 mg (23%) of Nl,N4-bis(l-(3- (6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolm-4-yl)phenylamino)-l-oxo-5,8,l l- tnoxa-2-azatπdecan-13-yl)-2,3-dihydroxysuccmamide as a white solid 1H-NMR (400MHz, DMSO, ppm) δ 8 68 (s, 2H), 7 68 (s, 2H), 7 64 (t, 2H), 7 39 (s, 2H), 7 24- 7 28 (m, 6H), 6 77-6 78 (m, 4H), 6 23 (s, 2H), 447 (s, 4H), 4 23 (s, 2H), 3 76 (s, 4H), 3 42-3 69 (m, 24H), 3 28-3 36 (m, 4H), 3 20 3 24 (m, 6H), 3 02 (s, 6H) MS (m/z) 583 [1/2M+1]+
Example 175
Nl,N2-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)oxalamide
Figure imgf000290_0001
Intermediate 175.1, N-(2-(2-(2-aminoethoxy)ethoxy)ethyl)-4-(6,8-dichloro-2- methyH,2,3,4-tetrahydroisoquinolin-4-yI)benzenesulfonamide: To 4-(6,8-dichloro- 2-methyl- 1 ,2,3,4-tetrahydroisoquinolin-4-yl)benzene-l -sulfonyl chloπde hydrochloπde (intermediate 10 6) (9 g, 20 02 mmol, 1 00 equiv, 95%) in DCM (200 mL) was added 2-(2-(2-aminoethoxy)ethoxy)ethanamine (15 6 g, 105 41 mmol, 5 00 eqmv) and tnethylamme (4 26 g, 42 18 mmol, 2 00 eqmv) and the resulting solution was stirred for 3 h at room temperature The reaction mixture was diluted with 100 mL of DCM and then washed with 2x50 mL of Bπne The mixture was dried over anhydrous sodium sulfate and concentrated under vacuum The residue was applied onto a silica gel column with DCM/methanol (10 1) This resulted m 3 g (28%) of N-(2-(2-(2- aminoethoxy)ethoxy)ethyl)-4-(6,8-dichloro-2-methyl- 1 ,2,3 ,4-tetrahydroisoquinohn-4- yl)benzenesulfonamide as brown oil
Figure imgf000290_0002
Compound 175, Nl,N2-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)oxalamide:
Into a 50-mL round-bottom flask, was placed a solution of N-(2-(2-(2- aminoethoxy)ethoxy)ethyl)-4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqmnolm-4- yl)benzenesulfonamide (intermediate 175 1) (150 mg, 028 mmol, 2 50 equiv, 92%) in DMF (5 mL), bis(2,5-dioxopyrrohdm-l-yl) oxalate (34 mg, 0 12 mmol, 1 00 equiv) and tnethylaniine (49 mg, 0 49 mmol, 4 00 equiv) The resulting solution was stirred overnight at room temperature The crude product was purified by Prep-HPLC with acetomtπle water (005% CF3COOH) (10%-l 00%) This resulted in 97 mg (68%) of a TFA salt ofNl,N2-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolm- 4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)oxalamide as a white solid 1H-NMR (300MHz, CD3OD, ppm) δ 7 90 (m, 4H), 7 56 (s, 2H), 7 50 (m, 4H), 6 85 (s, 2H), 4 77 (m, 4H), 4 53 (d, 2H), 3 90(m, 2H), 3 88 (m, 10H), 3 58 (m, 12H), 3 31(s, 6H), 3 12 (m, 4H) MS (m/z) 530 [l/2M+l]+
Example 176
Nl,N4-bis(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethyl)-2,3-dihydroxysuccin amide
Figure imgf000291_0001
Intermediate 176.1, N-(2-(2-aminoethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide: Into a 50 mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 2- (2-aminoethoxy)ethanamine dihydrochloπde (1 0 g, 5 65 mmol, 5 52 equiv) in DMF (20 mL), potassium carbonate (2 0 g, 14 39 mmol, 14 05 equiv) and 3-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoquinolm-4-yl)benzene-l-sulfonyl chloπde (intermediate 1 6) (400 mg, 1 02 mmol, 1 00 equiv) The resulting solution was stirred overnight at room temperature at which time it was diluted with 100 mL of water The resulting solution was extracted with 3x30 mL of ethyl acetate and the organic layers were combined and dried over sodium sulfate and concentrated under vacuum This resulted in 60 mg (13%) of N-(2-(2-aminoethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolm-4-yl)benzenesulfonamide as a yellow solid
Figure imgf000292_0001
10 Compound 176, Nl,N4-bis(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethyl)-2,3- dihydroxysuccinamide: Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of N-(2-(2-ammoethoxy)ethyl)- 3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolm-4-yl)benzenesulfonamide
I5 (intermediate 176 1) (60 mg, 0 13 mmol, 1 00 equiv) in DMF (3 mL), bis(2,5- dioxopyrrolidin-1-yl) 2,3 dihydroxybutanedioate (intermediate 168 1) (21 mg, 0 06 mmol, 0 47 equiv) and tπethylamine (50 mg, 049 mmol, 3 77 equiv) The resulting solution was stirred overnight at room temperature at which time the mixture was concentrated under vacuum The crude product was purified by Prep-HPLC with
20 acetomtπle water (0 05% CF3COOH) (10%-100%) This resulted in 21 mg (13%) of a TFA salt of Nl,N4-bis(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolm-4- yl)phenylsulfonamido)ethoxy)ethyl)-2,3-dihydroxysuccmamide as a white solid 1H- NMR (300MHz, CD3OD, pprri) δ 7 92 (d, J=I 8Hz, 2H) , 7 81(m, 2H), 7 67 (t, J=I 8Hz, 2H) , 7 57(m, 2H), 7 55 (d, J=6 9Hz, 2H) , 6 85(m, 2H), 4 78(s, 2H) , 4 77(s, 2H) , 4.54(d, J=402Hz, 2H), 4.48(s, 2H), 3 92(m, 2H), 3 53(m, 10H), 3 18(s, 6H), 3 07(t, J=5 4Hz, 4H) MS (m/z) 517 [1/2M+1 ]+
Example 177
Nl,N4-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)succinamide
Figure imgf000293_0001
Intermediate 177.1, bis(2,5-dioxopyrrolidin-l-yl) succinate: To succinic acid (3 0 g, 25 42 mmol, 1 OO equiv) m THF (50 mL) was added a solution of 1- hydroxypyrrohdine-2,5-dione (64 g, 55 65 mmol, 2 20 eqmv) This was followed by the addition of a solution of DCC (11 5 g, 55 83 mmol, 2 20 equiv) in THF (50 mL) dropwise with stirring at O0C The resulting solution was stirred overnight at room temperature The reaction progress was monitored by LCMS The solids were collected by filtration and the filtrate was concentrated to give the crude product The resulting solids were washed with THF and ethanol This resulted in 2 4 g (27%) of bis(2,5- dioxopyrrohdin-1-yl) succinate as a white solid
Figure imgf000293_0002
Compound 177, Nl,N4-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)succinamide:
Compound 177 was prepared using the procedure descπbed m example 175, substituting (2,5-dioxopyrrohdin-l-yl) succinate (intermediate 177 1) for bis(2,5- dioxopyrrolidin-1-yl) oxalate The crude product was purified by Prep-HPLC with acetomtπle water (0 05% CF3COOH) (10%-100%) This resulted in 32 8 mg (8%) of Nl,N4-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l>2,3,4-tetrahydroisoquinolm-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)succmamide as a white solid 1H-NMR (300MHz, CD3OD, ppm) δ 7 93-7 91 (d, J-S IHz, 4H), 7 57-7 56 (d, J~\ 8Hz, 2H), 7 50-7 47 (d, J=S, 4Hz, 4H), 6 86 (s, 2H), 4 78-4 73 (d, J-13 5Hz, 4H), 4 52 (m, 2H), 3 85 (m , 2H), 3 59-3 47 (m, 18H), 3 15-3 09 (m, 10H), 2 49 (s, 4H) MS (m/z) 544 [1/2M+1]+
Example 178
2,2'-oxybis(N-(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)acetamide)
Figure imgf000294_0001
Intermediate 178.1, bis(2,5-dioxopyrrolidin-l-yl) 2,2'-oxydiacetate: Intermediate 178 1 was prepared using the procedure outlined in example 177, substituting 2,2'- oxydiacetic acid for succinic acid The crude product was washed with ethyl acetate This resulted in 1 5 g (19%) of Intermediate 178 1 as a white solid
Figure imgf000295_0001
Compound 178, 2,2'-oxybis(N-(2-(2-(2-(4-(6,8-dichIoro-2-inethyl-l,2,3,4- tetrahydroisoquinoIin-4-yI)phenylsulfonamido)ethoxy)ethoxy)ethyl)acetainide):
Compound 178 was prepared using the procedure described in example 175, substituting bis(2,5-dioxopyrrolidin-l-yl) 2,2'-oxydiacetate (intermediate 178.1) for bis(2,5-dioxopyrrolidin-l-yl) oxalate. The crude product was purified by Prep-HPLC with acetonitrile:water (0.05% CF3COOH) (10%-100%). This resulted in 39.1 mg (7%) of a TFA salt of 2,2'-oxybis(N-(2-(2-(2-(4-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)acetamide) as a white solid. 1H-NMR (300MHz, CD3OD1^m): δ 7.94-7.91(m, 4H), 7.57-7.56(m, 2H), 7.51-7.48(m, 4H), 6.87(m, 2H), 4.82-4.76(m, 4H), 4.54-4.49(m, 2H), 3.93-3.91(s, 4H), 3.89-3.87(m, 2H), 3.66-3.42(m, 22H), 3.17(s, 6H), 3.13-3.09(m, 4H).MS {m/z): 552 [1/2M+1]+.
Example 179
(2R,3R)-Nl,N4-bis(2-(2-(2-(3-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylamino)-3-oxopropoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide
H0^-o-^σ^-'0H ^ o n I THRNa(cat)
Intermediate 179.1, tert-butyl 3-(2-(2-(2- hydroxyethoxy)ethoxy)ethoxy)propanoate: To triethyleneglycol (17.6 g, 117.20 mmol, 3 00 equiv) in anhydrous THF (70 mL), was added sodium (30 mg, 1 25 mmol, 0 03 equiv) Tert-butyl acrylate (5 0 g, 39 01 mmol, 1 00 eqmv) was added after the sodium had dissolved The resulting solution was stirred overnight at room temperature and then neutralized with I O N hydrogen chloride After removal of the solvent, the residue was suspended in 50 mL of bπne and extracted with 3x50 mL of ethyl acetate The combined organic layers were washed with saturated bnne and dπed over anhydrous sodium sulfate After evaporation of the solvent, the tert-butyl 3-(2-(2-(2- hydroxyethoxy)ethoxy)ethoxy)propanoate (9 6 g) was isolated as a colorless oil, which was used directly for the next reaction step without further purification
Figure imgf000296_0001
Intermediate 179.2, tert-butyl 3-(2-(2-(2-
(tosyloxy)ethoxy)ethoxy)ethoxy)propanoate Into a 250-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of tert-butyl 3-(2-(2 (2-hydroxyethoxy)ethoxy)ethoxy)propanoate (intermediate 179 1) (9 6 g, 3449 mmol, 1 00 equiv) m anhydrous pyridine (12 mL) The mixture was cooled to O0C and 4-methylbenzene-l-sulfonyl chloπde (7 9 g, 41 44 mmol, 1 20 equiv) was added slowly m several portions The resulting solution was stirred at O0C for 1-2 h and then the flask containing the reaction mixture was sealed and placed in a refrigerator at O0C overnight The mixture was poured into 120 mL of water/ice, and the aqueous layer was extracted with 3x50 mL of DCM The combined organic layers were washed with 2x50 mL of cold I O N hydrogen chloπde and saturated bnne and dπed over anhydrous sodium sulfate The solvent was removed under vacuum to yield 13 4 g (90%) of tert-butyl 3-(2-(2-(2-(tosyloxy)ethoxy)ethoxy)ethoxy)propanoate as pale yellow oil
Figure imgf000296_0002
Intermediate 179.3, tert-butyl 3-(2-(2-(2-(l,3-dioxoisoindolin-2- yl)ethoxy)ethoxy)ethoxy)propanoate: Into a 250-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of tert-butyl 3- (2-(2-(2-(tosyloxy)ethoxy)ethoxy)ethoxy)propanoate (13 4 g, 30 98 mmol, 1 00 eqmv) m anhydrous DMF (100 mL) followed by potassium phthalimide (7 5 g, 4049 mmol, 1 31 equiv) The resulting solution was heated to 1000C and stirred for 3 h The reaction progress was monitored by LCMS The DMF was removed under vacuum to afford a brown oil residue To the residue was added 200 mL water and the mixture was extracted with 3x50 mL of ethyl acetate The combined organic layers were washed with saturated bπne and dned over anhydrous sodium sulfate After evaporation of solvent, The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (0~l 3) The solvent was removed from fractions containing phthalimide and the residue was washed with 20% ethyl acetate/petroleum ether to yield 10 1 g (78%) of tert-butyl 3-(2-(2-(2-(l ,3-dioxoisoindolin-2-yl)ethoxy)ethoxy)ethoxy)propanoate as pale yellow oil
Figure imgf000297_0001
Intermediate 179.4, 3-(2-(2-(2-(l,3-dioxoisoindolin-2- yl)ethoxy)ethoxy)ethoxy)propanoic acid: Into a 10-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of tert-butyl 3-(2-(2 (2 (l,3-dioxoisoindolin-2-yl)ethoxy)ethoxy)ethoxy)propanoate (intermediate 179 3) (1 5 g, 3 68 mmol, 1 00 equiv) in neat 2,2,2 tπfluoroacetic acid (TFA, 2 0 mL) The resulting solution was stirred for 40 min at ambient temperature Excess TFA was removed under vacuum to afford a pale yellow oil residue which was purified on a silica gel column eluting with ethyl acetate/petroleum ether (1 5~1 2~2 1) to yeild 1 1 g (84%) of 3-(2-(2-(2-(l,3-dioxoisoindolm-2-yl)ethoxy)ethoxy)ethoxy)propanoic acid as a white solid
Figure imgf000298_0001
Intermediate 179.5, 3-(2-(2-(2-(l,3-dioxoisoindolin-2- yl)ethoxy)ethoxy)ethoxy)propanoyl chloride: Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 3- (2-(2-(2-(l,3-dioxoisoindolm-2-yl)ethoxy)ethoxy)ethoxy)propanoic acid (700 mg, 1 99 mmol, 1 00 equiv) in anhydrous DCM (300 mL), then oxalyl dichloπde (0 7 mL) was added dropwise at room temperature Two drops of anhydrous DMFwere then added The resulting solution was heated to reflux for 40 mm The solvent was removed under vacuum to yield 750 mg of 3-(2-(2-(2-(l,3-dioxoisomdohn-2- yl)ethoxy)ethoxy)ethoxy)ρropanoyl chloπde as pale yellow oil, which was used directly for the next reaction step without further purification
Figure imgf000298_0002
Intermediate 179.6, N-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenyl)-3-(2-(2-(2-(l,3-dioxoisoindolin-2-yl)ethoxy)ethoxy)ethoxy)propanamide:
To 3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolm-4-yl)benzenamine
(intermediate 31 5) (600 0 mg, 1 95 mmol, 1 00 equiv) in anhydrous DCM (5 0 mL) was added N-ethyl-N,N-dnsopropylamine (DIEA, 0 5 mL) Then a solution of 3-(2-(2- (2-(l ,3-dioxoisoindolm-2-yl)ethoxy)ethoxy)ethoxy)propanoyl chloπde (intermediate 179 5) (794 mg, 2 15 mmol, 1 10 equiv) was added dropwise with stirring at room temperature The resulting solution was stirred for 2 h at ambient temperature and then concentrated under vacuum The residue was applied onto a silica gel column with DCM/methanol (100-50 1) This resulted in 870 mg (66%) of N-(3-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoqumohn-4-yl)phenyl)-3-(2-(2-(2-(l,3-dioxoisomdolin-2-
29δ yl)ethoxy)ethoxy)ethoxy)propanamide as a pale yellow syrup The other fractions was collected and evaporated to get an additional 200 mg of impure product
Figure imgf000299_0001
Intermediate 179.7, 3-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)-N-(3-(6,8-dichloro-2- methy]-l,2,3,4-tetrahydroisoquinolin-4-yl)phenyl)propanamide: Into a 100-mL round-bottom flask, was placed N-(3-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoquinolm-4-yl)phenyl)-3-(2-(2-(2-(l,3-dioxoisoindohn 2 yl)ethoxy)ethoxy)ethoxy)propanarmde (870 0 mg, 1 36 mmol, 1 00 eqmv) and IM hydrazine monohydrate in ethanol (30 0 mL, 30 0 mmol) The resulting solution was heated at reflux for 1 hour The resulting mixture was cooled to room temperature and concentrated under vacuum The residual solution was diluted with 30 mL of water and then extracted with 3x50 mL of DCM The combined organic layers were washed with bπne, dπed over anhydrous sodium sulfate and concentrated under vacuum The residue was applied onto a silica gel column with DCM/methanol (100-50 1-10 1-1 1) This resulted in 600 mg (85%) of 3-(2-(2-(2- aminoethoxy)ethoxy)ethoxy)-N-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoqumolm-4-yl)phenyl)propanamide as a pale yellow syrup
Figure imgf000300_0001
Compound 179, (2R,3R)-Nl,N4-bis(2-(2-(2-(3-(3-(6,8-dichloro-2-methyI-l,2,3,4- tetrahydroisoquinolin-4-yl)phenyIamino)-3-oxopropoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide: To 3-(2-(2-(2-ammoethoxy)ethoxy)ethoxy)-N-(3-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolin-4-yl)phenyl)propanamide (intermediate 179 7) (270 mg, 0 53 mmol, 2 00 eqmv) in anhydrous DMF (5 0 mL) was added (2R,3R)-bis(2,5-dioxopyrrohdm-l-yl) 2,3-dihydroxysuccinate (prepared from (2R,3R)-tartaπc acid as descπbed in example 168) (91 0 mg, 0 26 mmol, 1 00 equiv) and tπethylamme (0 3 mL) and the resulting solution was stirred for 2 h at 350C The resulting mixture was then concentrated under vacuum The residue was purified by Prep-HPLC, to give 170 mg (56%) of a TFA salt of (2R,3R) Nl, N4 bis(2-(2-(2-(3-(3- (6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumohn-4-yl)ρhenylammo)-3 oxopropoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide as an off-white solid 1H NMR (300MHz, CD3OD, ppm) δ 7 92 (s, IH), 7 65 (s, 2H), 7 54 (d, J=I 5Hz, 2H), 7 36-7 46 (m, 4H), 7 02 (dd, J=I 5, 1 2Hz, 2H), 6 90 (s, 2H), 4 83 4 75 (m, 2H), 4 65- 4 60 (m, 2H), 4 53 (s, IH), 4 46 (m, 3H), 3 88-3 80 (m, 6H), 3 64 3 51 (m, 22H), 3 41- 3 35 (m, 4H), 3 16 (s, 6H), 2 64 (t, J~6 OHz, 4H) MS (m/z) 1136 [M+H]+
Example 180
Nl,N2-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinoIin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)oxalamide
Figure imgf000301_0001
Compound 180, Nl,N2-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoqumo]in-4-yl)pheny]suIfonamido)ethoxy)ethoxy)ethoxy)- 5 ethyl)oxalamide: Compound 180 was prepared from compound 28 following the procedure outlined in example 175 The crude product (400 mg) was puπfied by Flash- Prep-HPLC with the following conditions Column, C18 silica gel, mobile phase, CH3CN/H2O/CF3COOH=39/100/0 05 increasing to
CH3CN/H2O/CF3COOH=39/100/0 05 within mm, Detector, UV 254 nm This resulted
10 in 113 4 mg (11%) of a TFA salt of Nl,N2-bis(2 (2-(2-(2-(3-(6,8-dichloro-2-methyl- 1 ,2,3,4-tetrahydroisoquinolm-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)oxalamide as a white solid 1H- NMR (300MHz, DMSO+DC1, ppm) δ 7 766(d, J=I 5Hz, 2H), 7 683(s, 2H), 7 586~7 637(m, 4H), 7 537(d, J-7 8Hz, 2H), 6 644(s, 2H), 4 834-4 889(m, 2H),
15 4 598(d, J=16 2Hz, 2H), 4 446(d, J=15 OHz, 2H), 3 602-3 763(m, 4H), 3 299-3 436(m, 24H), 3 224-3 263(m, 4H), 2 975(s, 6H), 2 825-2 863(m, 4H) MS (m/z) 574 [M/2+H]+
Example 181
20 Nl,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)succinamide
Figure imgf000302_0001
Compound 181, Nl,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-
5 yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)succinamide: Compound 181 was prepared from compound 28 and (2,5-dioxopyrrolidin-l-yl) succinate following the procedure outlined in example 175 The crude product (200 mg) was purified by Flash- Prep-HPLC with the following conditions Column, Cl 8 silica gel, mobile phase, CH3CN/H2O/CF3COOH=0 05/100/0 05 increasing to
10 CH3CN/H2O/CF3COOH=90/100/0 05 within 19 mm, Detector, UV 254 nm This resulted m 201 mg (78%) of a TFA salt of Nl,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2- methyl- 1 ,2,3 ,4-tetrahydroisoquinolm-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)succinamide as a white solid 1H- NMR (300MHz, DMSO+DC1, ppm) δ 7 76(d, J=I 5Hz, 2H), 7 68(s, 2H),
15 7 63-7 52(m, 6H), 6 64(s, IH), 4 88-4 82(m, 2H), 4 62-4 42(m, 4H), 3 76-3 60(m, 4H), 3 43-3 30(m, 25H), 3 14-3 10(m, 4H), 2 97(s, 6H), 2 86-2 82(m, 4H), 2 27(s, 4H) MS (m/z) 589 [M/2+l]+
Example 182
20 Nl,N3-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyJ)-2,2-dimethylmalonamide
Figure imgf000303_0001
Figure imgf000303_0002
Compound 182, Nl,N3-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonainido)ethoxy)ethoxy)ethoxy)ethyl)-2,2- 5 dimethylmalonamide: Compound 182 was prepared from compound 28 and bis(2,5- dioxopyrrolidin-1-yl) 2,2-dimethylmalonate (prepared using the methods outlined m example 168) following the procedure outlined in example 175 The crude product (250 mg) was purified by Prep-HPLC with the following conditions Column, C18 sihca gel, mobile phase, MeCN/H2O/CF3COOH-39/100/0 05, Detector, UV 254 nm
10 This resulted in 152 3 mg (47%) of a TFA salt of Nl,N3-bis(2-(2-(2-(2-(3-(6,8- dichloro-2 -methyl- 1 ,2,3 ,4-tetrahydroisoquinohn-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,2-dimethylmalonamide as a white solid 1H-NMR (SOOMHz1 CDCl31 ^Pm) δ 7 92-7 89(d, J=S IHz, 2H), 7 79 (s, 2H), 7 69-7 64 (m, 2H), 7 57-7 55 (d, 7=7 5Hz, 4H), 3 68 (s, 2H), 4 87-4 75 (m, 4H),
15 4 54-4 49 (m, 2H), 3 90-3 88 (m ,2H), 3 67-3 45 (m, 20H), 3 39-3 32 (m, 4H) ,3 31 (s, 6H), 3 17-3 05 (m, 4H), 1 41(s,lH) MS (m/z) 1189 [M+H]+
Example 183
Nl,N3-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- 20 yl)phenyIsulfonamido)ethoxy)ethoxy)ethyl)-2,2-dimethylmalonamide
Figure imgf000304_0001
Example 183, Nl,N3-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-2,2- dimethylmalonamide: Compound 183 was prepared from intermediate 175 1 and bis(2,5-dioxopyrrohdm-l-yl) 2,2-dimethylmalonate (prepared using the methods outlined in example 168) following the procedure outlined in example 175 The crude product was purified by Prep-HPLC with acetomtnle water (0 05% CF3COOH)(10%- 100%) This resulted in 29 5 mg (5%) of a TFA salt of Nl,N3-bis(2-(2-(2-(4-(6,8- dichloro-2-methyl-l ,2,3,4-tetrahydroisoquinolm-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-2,2-dimethylmalonamide as a white solid 1H-NMR (300MHz, CD3OD, ppm) δ 7 94-7 92(m, 4H), 7 57(m, 2H), 7 51-7 49(m, 4H), 6 87(m, 2H), 4 83-4 74(m, 4H), 4 55-4 50(m, 2H), 3 92-3 87(m, 2H), 3 67-3 48(m, 8H), 3 40-3 38(m, 4H), 3 18(s, 6H), 3 14-3 00(m, 4H), 1 41(s, 6H) MS (m/z) 551 [1/2M+H]+
Example 184
N,N'-(2,21-(2,2'-(2,21-(2,2'-(pyridlne-2,6-diylbis(oxy))bls(ethane-2,l- diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane- 2,l-diyl))bis(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)benzenesulfonamide) TsCl
HCX .,OTs
Et3N, DCM, rt w w
Intermediate 184.1, 2-(2-(2-(2-hydroxyethoxy)ethoxy)ethoxy)ethyl 4- methylbenzenesulfonate: Into a 250-mL round-bottom flask was placed a solution of tetraethylene glycol (50 g, 257 47 mmol, 9 81 equiv) m DCM (150 mL) and tnethylamme (8 g, 79 05 mmol, 3 01 equiv) This was followed by the addition of a solution of 4-methylbenzene-l-sulfonyl chloπde (5 0 g, 2623 mmol, 1 00 equiv) m DCM (10 mL) dropwise with stirring at O0C The resulting solution was stirred for 2 h at room temperature, at which time it was diluted with 200 ml of hydrogen chlonde(3N aq ) The resulting solution was extracted with 2x150 mL of DCM and the combined organic layers were washed with 3x150 mL of saturated sodium bicarbonate The mixture was dried over sodium sulfate and concentrated under vacuum The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 5- ethyl acetate) This resulted in 7 0 g (77%) of 2 (2 (2 (2-hydroxyethoxy)ethoxy)ethoxy)ethyl 4-methylbenzenesulfonate as colorless oil
0Ts NaN3 NaHCO3
DMF 80
Intermediate 184.2, 2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethanol: To intermediate 184 1 (2 0 g, 5 74 mmol, 1 00 equiv) m DMF (40 mL) was added sodium azide (700 mg, 10 77 mmol, 1 88 equiv) and sodium bicarbonate (800 mg, 9 52 mmol, 1 66 equiv) The resulting solution was stirred for 2 h at 8O0C at which time the mixture was concentrated under vacuum The residue was diluted with 100 mL of water and then extracted with 3x100 mL of DCM The organic layers were combined and concentrated under vacuum to afford 1 3 g of 2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethanol as light yellow oil Br N Br
HO^/-^
NaH DMF 80
Figure imgf000306_0001
Intermediate 184.3, 2,6-bis(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethoxy)pyridine
Into a 50-mL round-bottom flask, was placed a solution of intermediate 1842 (220 mg, 1 00 mmol, 2 38 eqmv) in DMF (10 mL) and sodium hydπde (40 mg, 1 00 mmol, 2 37 eqmv, 60%) The resulting solution was stirred for 30 mm at room temperature, at which time 2,6-dibromopyπdine (100 mg, 0 42 mmol, 1 00 eqmv) was added The resulting solution was stirred for an additional 2 h at 8O0C, and then was concentrated under vacuum The residue was applied onto a silica gel column with DCM/methanol (50 1-30 1) This resulted in 180 mg (83%) of 2,6-bis(2-(2-(2-(2- azidoefhoxy)efhoxy)ethoxy)ethoxy)pyridine as light yellow oil
Figure imgf000306_0002
Intermediate 184.4, 2-(2-(2-(2-(6-(2-(2-(2-(2- aminoethoxy)ethoxy)ethoxy)ethoxy)pyridin-2- yloxy)ethoxy)ethoxy)ethoxy)ethanamine: To intermediate 184 3 (180 mg, 035 mmol, 1 00 eqmv) in THF/water (30/3 mL) was added tπphenylphosphine (400 mg, 1 52 mmol, 4 35 equiv) and the resulting solution was stirred overnight at 4O0C Aftercooling to room temperature, the reaction mixture was extracted with 4x50 mL of DCM and the organic layers combined and dπed over anhydrous sodium sulfate and concentrated under vacuum The residue was applied onto a silica gel column with DCM/methanol (80 1-20 1) This resulted in 100 mg (62%) of 2-(2-(2-(2-(6-(2-(2-(2- (2-aminoethoxy)ethoxy)ethoxy)ethoxy)ρyndin-2- yloxy)ethoxy)ethoxy)ethoxy)ethanamme as light yellow oil
Figure imgf000307_0001
Compound 184, N,N'-(2,2'-(2,2'-(2,2'-(2,2'-(pyridine-2,6-diylbis(oxy))bis(ethane- 2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-
10 diyl))bis(oxy)bis(ethane-2,l-diyI))bis(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yt)benzenesulfonamide): To intermediate 1844 (100 mg, 0 22 mmol, 1 00 equiv) in DCM (50 mL) was added tπethylamine (70 mg, 0 69 mmol, 3 20 equiv) and 3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolm-4-yl)benzene-l- sulfonyl chloride (350 mg, 0 90 mmol, 4 13 equiv) The resulting solution was stirred
I5 overnight at room temperature, and then concentrated under vacuum The residue was purified by Prep HPLC with CH3CN H2O(O 05% CF3COOH)=35% 40% This resulted m 88 4 mg (29%) of a TFA salt of the title compound as a white solid 1H-NMR (300MHz, CD3OD, pprri) δ 7 91-7 88(d, 2H), 7 78(s, 2H), 7 67-7 50(m, 7H), 6 86(s, 2H), 6 34-6 31(d, 2H), 4 90-4 75 (m, 4H), 4 52-4 46(m, 2H), 442-4 39(t, 4H), 3 90-
20 3 81(m, 6H), 3 71-3 43 (m, 22H), 3 16(s, 6H), 3 07-3 03(t, 4H) MS (m/z) 1170 [M+H]+
Example 185
3O5 2,2Mmethylazanediyl)bis(TH2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)acetamide) tris(2,2,2-trifluoroacetate)
O^NwO O O
O ' O n DrCrC/™THCF VQ ° ' ° (/~~/
Intermediate 185.1, bis(2,5-dioxopyrro)idin-l-yl) 2,2'-(methylazanediy])diacetate
To 2-[(carboxymethyl)(methyl)amino]acetic acid(2 0 g, 13 60 mmol, 1 00 equiv) in THF (30 mL) was added DCC (62 g, 30 05 mmol, 2 21 eqmv) and a solution of NHS (3 5 g, 30 41 mmol, 2 24 eqmv) m THF (30 mL) and the reaction stirred at 0-100C for 2 h The resulting solution was allowed to warm to room temperature and stirred for 16 h The solids were then filtered out, and the resulting mixture was concentrated under vacuum The crude product was re-crystallized from ethyl acetate/petroleum ether in the ratio of 1 10 to afford 2 0 g (21%) of the title compound as a white solid
Figure imgf000308_0001
Compound 185, 2,2'-(methylazanediyl)bis(N-(2-(2-(2-(4-(6,8-dichloro-2-methyl- l,2,3,4-tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)- acetamide) tris(2,2,2-trifluoroacetate) To N-(2-(2-(2-aminoethoxy)ethoxy)ethyl)-4- (6,8 dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin 4-yl)benzenesulfonamide (150 mg, 0 30 mmol, 1 00 equiv) in DMF (3 mL) was added intermediate 185 1 (106 mg, 0 15 mmol, 0 50 equiv, 48%) and tπethylamme (150 mg, 1 48 mmol, 4 97 equiv) and the reaction was stirred overnight The mixture was concentrated under vacuum and the crude product was punfied by Prep-HPLC with CH3CN H2O (0 05% CF3COOH) to afford 26 4 mg (12%) of a TFA salt of the title compound as a white solid 1H-NMR (300MHz, CD3OD, ppm) 5 7 92 (m, 4H), 7 5 (m, 2H), 7 50 (m, 4H), 6 85 (s, 2H), 4 81 (m, 4H), 4 50 (m, 2H), 4 06 (s, 4H), 3 89 (m, 2H), 3 66-3 44 (m, 22H), 3 32 (s, 6H), 3 15 (m, 4H), 3 01 (s, 3H) MS (m/z) 559 [(M+2H)/2] +
Example 186 5-amino-Nl,N3-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)isophthalamide tris(2,2,2- trifluoroacetate)
Figure imgf000309_0001
Intermediate 186.1, bis(2,5-dioxopyrrolidin-l-yl) 5-aminoisophthalate: Into a 50- mL 3-necked round-bottom flask, was placed a solution of 5-aminoisophthalic acid (300 mg, 1 66 mmol, 1 00 equiv) in THF (5 mL) and l-hydroxypyrrolidme-2,5-dione (420 mg, 3 65 mmol, 2 20 equiv) This was followed by the addition of a solution of DCC (750 mg, 3 64 mmol, 2 20 equiv) in THF (5 mL) dropwise with stimng at O0C The resulting solution was stirred overnight at room temperature The solids were removed by filtration and the filtrate was concentrated under vacuum The crude product was punfied by re-crystallization from ethanol This resulted in 70 mg (11 %) of the title compound as a light yellow solid
Figure imgf000310_0001
Compound 186, 5-amino-Nl,N3-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)isophthalamide tris(2,2,2- trifluoroacetate): To N-(2-(2-(2-aminoethoxy)ethoxy)ethyl)-4 (6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoqumohn-4-yl)benzenesulfonamide (100 mg, 0 20 mmol, 1 00 equiv) in DMF (5 mL) was added intermediate 186 1 (44 8 mg, 0 12 mmol, 060 equiv) and tnethylamine (604 mg, 0 60 mmol, 3 00 equiv) and the reaction was stirred overnight The resulting mixture was concentrated under vacuum and the crude product was punfied by Prep-HPLC with CH3CN H2O (0 05% CF3COOH) to afford 32 4 mg (19%) of a TFA salt of the title compound as a white solid 1H-NMR (300MHz, CD3OD, ppm) δ 7 90-7 87 (d, JS 4Hz, 4H), 7 60-7 54 (3H, m), 7 46-7 44(d, J=B 4Hz, 4H), 7 34 (d, J 1 2Hz, 2H), 6 82 (s, 2H), 4 89-4 71 (m, 4H), 4 53-448 (d, >16 2Hz, 2H), 3 91-3 85 (m, 2H), 3 67-3 45 (m, 22H), 3 33-3 32 (m, 6H), 3 18-3 01 (m, 4H) MS (m/z) 575 [(M+2H)/2] +
Example 187 2,2'-oxybis(N-(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethy])acetamide)
Figure imgf000311_0001
Compound 187, 2,2'-oxybis(N-(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)acetamide): Into a 50-mL round- bottom flask, was placed a solution of N-(2-(2-(2-(2-
10 aminoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l ,2,3,4- tetrahydroisoqumolm-4-yl)benzenesulfonamide (compound 28) (150 mg, 028 mmol, 1 00 equiv) in DMF(5 mL), tπethylamine (56 mg, 0 55 mmol, 2 01 eqmv) and bis(2,5- dioxopyrrolidin-1-yl) 2,2'-oxydiacetate (intermediate 178 1) (44 mg, 0 14 mmol, 0 49 equiv) The resulting solution was stirred overnight at room temperature, at which time
I5 the mixture was concentrated under vacuum The crude product (150 mg) was purified by preparative HPLC with the following conditions Column, Cl 8 silica gel, mobile phase, methanol/water = 0 05/100 increasing to methanol/water = 90/100 withm 19 mm, Detector, UV 254 trni This resulted in 72 4 mg (44%) of the title compound as a white solid 1H NMR (300MHz, CD3OD, ppm) δ 7 79 (d, J=I 2Hz, 2H), 7 71 (s, 2H),
20 7 49-7 58 (m, 4H), 7 36-7 37 (m, 2H), 6 82 (s, 2H), 4 39-444 (m, 2H), 4 06 (s, 4H), 3 80 (d, J=162Hz, 2H), 3 65 (d, /=16 2Hz, 2H), 3 55-3 61 (m, 16H), 3 43-3 52 (m, 12H), 3 02-3 08 (m, 6H)1 2 65-2 70 (m, 2H), 2 49 (s, 6H) MS (m/z) 1190 [M+H] +
Example 188 5-bromo-Nl,N3-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)isophthalamide bis(2,2,2- trifluoroacetate)
Figure imgf000312_0001
Intermediate 188.1, 5-bromoisophthalic acid: Into a 100-mL round-bottom flask, was placed a solution of isophthalic acid (10 g, 60 24 mmol, 1 00 equiv) in 980ZoH2SO4 (60 mL) This was followed by the addition of N-bromosuccimmide (12 80 g, 72 32 mmol, 1 20 equiv), in portions at 6O0C in 10 mm The resulting solution was stirred overnight at 6O0C in an oil bath The reaction was cooled to room temperature and then quenched by the addition of water/ice The solids were collected by filtration, and washed with 2x60 mL of hexane The solid was dried in an oven under reduced pressure The crude product was purified by re-crystallization from ethyl acetate to give 3 g (20%) of 5-bromoisophthalic acid as a white solid
Figure imgf000312_0002
Intermediate 188.2, bis(2,5-dioxopyrrolidin-l-yl) 5-bromoisophthaIate Into a 100- mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 5-bromoisophthalic acid (3 g, 11 76 mmol, 1 00 equiv, 96%) in THF (20 mL) followed by NHS (3 g, 2609 mmol, 220 equiv) at 0-50C To this was added a solution of DCC (5 6 g, 27 18 mmol, 2 20 equiv) m THF (20 mL) dropwise with stirring at 0-50C The resulting solution was stirred overnight at room temperature The solids were filtered out and the filtrate was concentrated under vacuum The crude product was re-crystalhzed from DCM/ethanol in the ratio of 1 10 This resulted m 4 g (75%) of the title compound as a white solid
Figure imgf000313_0001
Compound 188, 5-bromo-Nl,N3-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinoIin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)isophthalamide bis(2,2,2- trifluoroacetate: Into a 50-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of N-(2-(2-(2- ammoethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolm-4- yl)benzenesulfonamide (intermediate 175 1) (100 mg, 0 19 mmol, 2 50 equiv, 95%) in DMF (8 mL), intermediate 188 1 (35 mg, 0 08 mmol, 1 00 equiv, 98%) and tπethylamine (32 mg, 0 32 mmol, 4 00 equiv) The resulting solution was stirred overnight at room temperature and then concentrated to dryness The crude product was purified by Prep-HPLC with acetomtπle water (0 05%CF3COOH) = 30%~42% This resulted in 86 mg (75%) of a TFA salt of the title compound as a white solid 1H-NMR (300 MHz, CD3OD, ppm) δ 826 (s, IH), 8 13 (s, 2H), 7 90 (d, J"9Hz, 4H), 7 55 (s, 2H), 748 (d, /=9Hz, 4H), 6 84 (s, 2H), 4 76 (m, 4H), 4 54 (m, 2H), 3 89 (m, 2H), 3 68 (m, 18H), 3 53 (m, 4H), 3 33 (s, 6H), 3 18 (m, 4H) MS (m/z) 609 [(M+2H)/2] +
Example 189
Nl,N3-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-2-hydroxymalonamide bis(2,2,2- trifluoroacetate)
Figure imgf000314_0001
Intermediate 189.1, bis(2,5-dioxopyrrolidin-l-yl) 2-hydroxymalonate Into a 100 ml 3 -necked roundbottom flask purged and maintained with an inert atmosphere of nitrogen, was placed a solution of 2-hydroxymalomc acid (1 6 g, 13 32 mmol, 1 00 equiv) in THF (30 niL) and DCC (6 2 g, 30 05 mmol, 2 26 equiv) This was followed by the addition of a solution of NHS (3 5 g, 30 41 mmol, 2 28 equiv) in THF (30 mL) at 0-100C m 2 h The resulting solution was stirred for 16 h at room temperature The solids were then filtered out and the filtrate was concentrated under vacuum The crude product was re-crystallized from ethanol to give 0 5 g (12%) of the title compound as a white solid
Figure imgf000315_0001
Compound 189, Nl,N3-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenyIsu]fonamido)ethoxy)ethoxy)ethyl)-2- hydroxymalonamide bis(2,2,2-trifluoroacetate): To N-(2-(2-(2- ammoethoxy)ethoxy)ethyl)-4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolm-4- yl)benzenesulfonamide (intermediate 175 1) (100 mg, 0 20 mmol, 1 00 eqmv) in DMF (2 mL), was added Intermediate 189 1 (29 mg, 0 10 mmol, 045 equiv) and tπethylamine (90 mg, 4 50 eqmv) and the reaction was stirred for 3 h at 300C The
10 mixture was concentrated under vacuum and the crude product was purified by Prep- HPLC with acetomtπle water (0 05% CF3COOH) (10%-100%) to afford 36 5 mg (30%) of a TFA salt of the title compound as a white solid 1H-NMR (300MHz, CD3OD, ppm) δ 7 94-7 91 (m, 4H), 7 57-7 56 (m, 2H), 7 51 7 48 (m, 4H), 6 87 (m, 2H), 4 82-4 76 (m, 4H), 4 54-449 (m, 2H), 3 93-3 91 (s, 4H), 3 89-3 87 (m, 2H), 3 66-
15 3 42 (m, 22H), 3 17 (s, 6H), 3 13-3 09 (m, 4H) MS (m/z) 546 [(M+2H)/2] +
Example 190
Nl,N2-bis(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsuLfonamido)ethoxy)ethoxy)ethyl)oxalamide
20
Figure imgf000316_0001
Compound 190, Nl,N2-bis(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)oxalamide: To N-(2-(2-(2-ammoemoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonarmde (Intermediate 168 2) (200 mg, 040 mmol, 1 00 equiv) in DMF (2 mL) was added tnethylamme (81 mg, 0 80 mmol, 2 01 equiv) and bis(2,5-dioxopyrrolidm-l-yl) oxalate (57 mg, 0 20 mmol, 0 50 equiv) and the resulting solution was stirred overnight The mixture was concentrated under vacuum and the crude product (200 mg) was purified by Flash-Prep-HPLC with the following conditions Column, Cl 8 silica gel, mobile phase, methanol/water=0 05/100 increasing to methanol/water=90/100 within 25 mm, Detector, UV 254 ran This resulted in 72 3 mg (34%) of Nl, N2-bis(2-(2-(2-(3-(6,8-dichloro-2-methyl- 1,2,3,4- tetrahydroisoquinohn-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)oxalamide as a light yellow solid 1H NMR (300MHz, CD3OD, ppm) δ 7 77-7 81 (m, 2H), 7 72 (s, 2H), 7 48-7 57 (m, 4H), 7 35-7 36 (m, 2H), 6 81-6 82 (m, 2H), 4 39-4 43 (m, 2H), 3 79 (d, /=16 5 Hz, 2H), 3 65 (d, J=16 2Hz, 2H), 3 55-3 60 (m, 8H), 3 43-3 50 (m, 12H), 3 02-3 09 (m, 6H), 2 64-2 71 (m, 2H), 2 49 (s, 6H) MS (m/z) 1059 [M+H]+
Example 191
Nl,N4-bis(2-(2-(2-(3-(6,8-dichIoro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)succinamide
Figure imgf000317_0001
Compound 191, Nl,N4-bis(2-(2-(2-(3-(6,8-dichIoro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsu]fonamido)ethoxy)ethoxy)ethyl)succinamide: To N-(2-(2-(2-aminoethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l ,2,3,4- tetrahydroisoqumolm 4 yl)benzenesulfonamide (intermediate 168 2) (150 mg, 0 30 mmol, 1 00 equiv) in DMF (2 mL) was added tπethylamme (60 mg, 0 59 mmol, 1 98 equiv) and intermediate 177 1 (47 mg, 0 15 mmol, 0 50 equiv) and the resulting solution was stirred overnight The mixture was then concentrated under vacuum and the crude product (150 mg) was purified by Flash-Prep-HPLC with the following conditions column, Cl 8 silica gel, mobile phase, methanol/water-0 05/100 increasing to methanol/water=90/100 within 25 mm, Detector, UV 254 run This resulted in 53 1 mg (33%) of Nl,N4-bis(2 (2-(2-(3-(6,8-dichloro 2 methyl- 1,2,3,4- tetrahydroisoqumolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)succinamide as a white solid 1H-NMR (300MHz, CD3OD, ppm) δ 7 77-7 80 (m, 2H), 7 71 (s, 2H), 7 48-7 57 (m, 4H), 7 36-7 37 (m, 2H), 6 82 (s, 2H), 4 39-4 44 (m, 2H), 3 79 (d, J-15 9Hz, 2H), 3 66 (d, J-16 2Hz, 2H), 3 45-3 57 (m, 16H), 3 35-3 37 (m, 4H), 3 03- 3 08 (m, 6H), 2 65-2 71 (m, 2H), 2 49-2 50 (m, 10H) MS (m/z) 1089 [M+H]+
Example 192
3,5-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinohn-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethylcarbamoyl)benzenesulfonic acid
Figure imgf000318_0001
Intermediate 192.1, sodium 3,5-bis((2,5-dioxopyrrolidin-l- yloxy)carbonyl)benzenesulfonate: To sodium 3,5-dicarboxybenzenesulfonate (1 g, 3 73 mmol, 1 00 eqmv) and NHS (940 mg, 8 17 mmol, 2 20 eqmv) in DMF (10 mL) at O0C was added dropwise a solution of DCC (1 69 g, 8 20 mmol, 2 20 equiv) m THF (10 mL) and the reaction stirred overnight The solids were removed by filtration and the filtrate was concentrated under vacuum to afford 500 mg (29%) of the title compound as a white solid
DMREt3N
Figure imgf000318_0002
Figure imgf000318_0003
Compound 192, 3,5-bis(2-(2-(2-(4-(6,8-dichIoro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl- carbamoyl)benzenesulfonic acid: To N-(2-(2-(2-ammoethoxy)ethoxy)ethyl)-4-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoquinohn-4-yl)benzenesulfonamide (intermediate 175 1) (100 mg, 0 20 mmol, 1 00 eqmv) in DMF (2 mL) was added intermediate 192 1 (45 mg, 0 10 mmol, 050 equiv) and tπethylamme (90 mg, 4 50 equiv) and the resulting solution was stirred overnight The mixture was concentrated under vacuum and the crude product was purified by Prep-HPLC with acetonitnle water (0 05% CF3COOH)(10%-100%) to afford 306 mg (22%) of a TFA salt of the title compound as a white solid 1H-NMR (300 MHz, CD3OD, ppm) δ 8 35- 8 34 (m, 3H), 7 84-7 81 (m, 4H), 7 48 (m, 2H), 7 41-7 38 (m, 4H), 6 75 (m, 2H), 4 87- 470 (m, 4H), 4 56-4 50 (m, 2H), 3 92-3 85 (m, 2H), 3 70-3 42 (m, 22H), 3 37-3 32 (m, 6H), 3 20-3 06 (m, 4H) MS (m/z) 608 [[(M+2H)/2]+
Example 193
Nl,N3-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethyI)-5-hydroxyisophthalamide
Figure imgf000319_0001
Intermediate 193.1, 5-hydroxyisophthalic acid: To dimethyl 5-hydroxyisophthalate (4 0 g, 19 03 mmol, 1 00 equiv) in THF (10 mL) was added lithium hydroxide (20 mL, 2M in water) and the resulting solution was stirred overnight at 4O0C The mixture concentrated under vacuum to remove the organic solvents and then the pH of the solution was adjusted to ~2 with 6N hydrochloric acid The resulting solids were collected by filtration and dπed in a vacuum oven to afford 2 O g (58%) of 5- hydroxyisophthahc acid as a white solid
Figure imgf000319_0002
Intermediate 193.2, bis(2,5-dioxopyrrolidin-l-yl) 5-hydroxyisophthalate: To 5- hydroxyisophthahc acid (Intermediate 193 1, 1 g, 5 49 mmol, 1 00 eqmv) and NHS (1 39 g, 220 equiv), m THF (5 mL) at O0C was added dropwise a solution of DCC (24 g, 2 20 equiv) in THF (5 mL) The resulting solution was stirred overnight at room temperature, then filtered and concentrated under vacuum to give 0 5 g (22%) of the title compound as a white solid
Figure imgf000320_0001
Compound 193, Nl,N3-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-5- hydroxyisophthalamide: To N-(2-(2-(2-aminoethoxy)ethoxy)ethyl)-4-(6,8-dichloro- 2-methyl-l,2>3,4-tetrahydroisoqumolin-4-yl)benzenesulfonamide (intermediate 175 1) (100 mg, 0 20 mmol, 1 00 equiv) m DMF (2 mL) was added Intermediate 193 2 (34 mg, 009 mmol, 045 equiv) and tnethylamine (90 mg, 4 50 equiv) and the reaction was stirred overnight The mixture was concentrated under vacuum and the crude product was puπfied by Prep-HPLC with acetonitrile water (0 05% CF3COOH)(10%-l 00%) to afford 30 mg (24%) of a TFA salt of the title compound as a white solid 1H-NMR (300MHz, CD3OD, ppm) δ 7 91-7 88 (m, 4H), 7 71-7 70 (m, IH), 7 56-7 55 (m, 2H), 7.47-7.44 (m, 4H), 7.37-7 36 (m, 2H), 6.84 (m, 2H), 4.87-4.70 (m, 4H), 4.53-448 (m, 2H), 3 92-3 85 (m, 2H), 3 67-3.46 (m, 22H), 3.37-3.32 (m, 6H), 3.17-3 07 (m, 4H). MS (m/z): 576 [[(M+2H)/2]+
S Example 194
(IRjSRVNl^-bistS-tCS^S^ό^-dichloro-l-methyl-l^^^-tetrahydroisoquinolin^- yl)phenylsulfonamido)propyl)(methyl)amino)propyl)-2,3-dihydroxysuccinamide
Figure imgf000321_0001
0
Intermediate 194.1, N-(3-((3-aminopropyl)(methyl)amino)propyl)-3-(6,8-dichloro- 2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonaniide: To a solution of Nl-(3-aminopropyl)-Nl-methyIpropane-l,3-diamme (560 mg, 3.85 mmol) dissolved in DCM (20 mL), was added tπethylamme (300 mg, 2 96 mmol) and 3-(6,8-dichloro-2-5 methyl-l,2,3,4-tetrahydroisoqumolm-4-yl)benzene-l-sulfonyl chloride (300 mg, 0.77 mmol). The resulting solution was stirred for 3 h at room temperature After removing the solvent, the resulting residue was diluted with EtOAc (50 mL), washed with water (2x10 mL) and dπed over anhydrous sodium sulfate. The crude product was purified by Flash-Prep-HPLC with H2O MeOH (1.4) to afford 300 mg (74%) of N-(3-((3-0 ammopropyl)(methyl)armno)propyl)-3-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoquinohn-4-yl)benzenesulfonamide as a yellow oil
Figure imgf000322_0001
Compound 194, (2R,3R)-Nl,N4-bis(3-((3-(3-(6,8-dichIoro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)propyl)(inethyl)aniino)propyl)-2,3- dihydroxysuccinamide: To a solution of N-(3-((3- aminopropyl)(methyl)ammo)propyl)-3-(6,8-dichloro-2-rnethyl-l, 2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide (Inteπnediate 194 1, 300 mg, 0 60 mmol) in DMF (2 mL) was added (2R,3R)-bis(2,5-dioxopyrrolidm-l-yl) 2,3- dihydroxysuccmate (prepared from (2R,3R)-tartaπc acid as descπbed in example 168) (91 mg, 0 27 mmol) and tπethylamme (270 mg, 2 67 mmol) and the resulting solution was stirred for 2 h at room temperature and the reaction progress was monitored by LCMS Upon completion, the mixture was concentrated under vacuum and the crude product was purified by Prep-HPLC with acetomtπle water (0 05% CF3COOH) (20%- 29%) to afford 30 9 mg (8%) of the title compound as a TFA salt 1H-NMR (300 MHz, CD3OD, ppm) 7 90-7 88 (m, 2H), 7 80 (m, 2H), 7 69-7 65 (m, 2H), 7 58-7 56 (m, 4H), 6 85 (m, 2H), 4 87-4 71 (m, 4H), 4 54-444 (m, 4H), 3 88-3 82 (m, 2H), 3 62-3 53 (m, 4H), 3 22 (m, 6H), 3 13-3 09 (m, 6H), 3 01-2 97 (m, 4H), 2 88 (m, 6H), 2 00-1 96 (m, 8H) LCMS (ES, m/z) 1114 [M+H]+
Example 195
2,2'-oxybis(N-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)acetamide)
Figure imgf000323_0001
Compound 195, 2,2'-oxybis(N-(2-(2-(2-(3-(6,8-dich]oro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonaιnido)ethoxy)ethoxy)ethyl)acetainide): To a solution of N-(2-(2-(2-ammoethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl- l,2,3,4-tetrahydroisoqumohn-4-yl)benzenesulfonamide (150 mg, 0.30 mmol) in DMF (2 mL) was added tπethylamme (60 mg, 0.59 mmol) and bis(2,5-dioxopyrrohdin-l-yl) 2,2'-oxydiacetate (intermediate 178 1) (49 mg, 0 15 mmol) and the resulting solution was stirred overnight After removal of the solvent, the crude product (150 mg) was puπfied by Flash-Prep-HPLC (C18 silica gel, methanol/water=0.05/100 increasing to methanol/water=90/100 within 25 mm) to give 44 4 mg (27%) of the title compound as a TFA salt. 1H-NMR (300 MHz, CD3CD, ppmf 7.79-7.76 (m, 2H), 7 70 (s, 2H), 7.57- 7 50 (m, 4H) , 7.36 (d, J=Hz, 2H), 4.89-4.41 (m, 2H), 4.06 (m, 4H), 3.81-3.62 (m, 5H), 3 59-3.42 (m, HH), 3 33-3 31 (m, 8H), 3 07-3 01 (m, 6H), 2 71-2 64 (m, 2H), 2 48(s, 6H). LCMS (ES, m/z): 1103[M+H]+.
Example 196
Nl,N3-bis(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-2,2-dimethylmalonamide
Figure imgf000324_0001
Compound 196, Nl,N3-bis(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenyIsu]fonamido)ethoxy)ethoxy)ethyl)-2,2- dimethylmalonamide: To N-(2-(2-(2-aminoethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoqumolin-4-yl)benzenesuIfonamide (150 mg, 0 30 mmol) m DMF (2 mL) was added tπethylamine (60 mg, 0 59 mmol) and bis(2,5-dioxopyrrolidin- 1-yl) 2,2-dimethylmalonate (prepared from 2,2-dimethylmalomc acid as descπbed in Example 168) (49 mg, 0 15 mmol) and the resulting solution was stirred overnight The mixture was concentrated and then purified by Flash-Prep-HPLC (C 18 silica gel, methanol/water=0 05/100 increasing to methanol/water=90/100 within 25 mm) to give 75 1 mg of the title compound (46%) as a TFA salt 1H-NMR (300MHz, CD3OD, ppiri) 7 80-7 77 (m, 2H), 7 71 (s,2H), 7 57-748 (m, 4H), 7 36-7 35 (d, J=I IHz, 2H), 6 81 (d, J=I 2Hz, 2H), 4 43-4 38 (m, 2H), 3 82-3 62 (m, 4H), 3 57—3 31 (m,18H), 3 07-3 02 (m, 6H), 2 71-2 64 (m, 2H), 2 49 (s, 6H), 1 41 (s, 6H) LC-MS (ES, m/z) 1101[M+H] +
Example 197
Nl,N2-bis(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)oxalamide
Figure imgf000325_0001
Compound 197, Nl,N2-bis(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)- ethyl)oxalamide: To a solution of N-(2-(2-(2-(2-ammoethoxy)ethoxy)ethoxy)ethyl)-4- (6,8-dichloro-2-memyl-l,2,3,4-tetrahydroisoquinolm-4-yl)benzenesulfonamide (compound 82) (148 mg, 0 26 mmol) in DMF (5 mL) under N2 was added bis(2,5- dioxopyrrolidin-1-yl) oxalate (prepared from oxalic acid as described in Example 168) (31 mg, 0.11 mmol) and tnethylamine (44 mg, 0.44 mmol) and the resulting solution was stirred overnight. The crude product was purified by Prep-HPLC with CH3CN:H2O(0.05%CF3COOH)(28%-35%) to afford 101.8 mg (68%) of the title compound as a TFA salt 1H-NMR (300Hz, CD3OD, ppm). 7 94 (d, J = 9Hz, 4H), 7 58 (s, 2H), 7 50 (d, J = 9Hz, 4H), 6.88 (s, 2H), 4 80 (m, 4H), 4.53 (m, 2H), 3.90 (m, 2H), 3 59 (m, 16H), 3 52 (m, 2H), 3.49 (m, 12H), 3.13 (s, 6H), 3.09 (m, 4H). LC-MS (ES, m/z) 574 [(M+2H)/2]+
Example 198
2,2'-oxybis(N-(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)acetamide)
Figure imgf000326_0001
Compound 198, 2,2'-oxybis(N-(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)acetamide): To a solution of N- (2-(2-(2-(2-ammoethoxy)emoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolm-4-yl)benzenesulfonamide (Compound 82) (200 mg, 0 37 mmol) in DMF (2 mL) was added bis(2,5-dioxopyrrolidm-l-yl) 2,2'-oxydiacetate (intermediate 178 1) (60 mg) and tπethylamme (184 mg) The resulting solution was stirred for 2 h at room temperature at which point LCMS indicated complete conversion The mixture was concentrated under vacuum and the crude product was purified by Prep-HPLC with acetomtπle water (0 05% CF3COOH)(25%-35%) This resulted in 79 6 mg (31%) of the title compound as a TFA salt 1H-NMR (300MHz, CD3OD, ppm) 7 94-7 91 (m, 4H), 7 58-7 57 (m, 2H), 7 51-7 48 (m, 4H), 6 88 (m, 2H), 4 82-4 74 (m, 4H), 4 52-4 47 (m, 2H), 4 06 (m, 4H), 3 90 (m, 2H), 3 64-3 42 (m, 34H), 3 15-3 13 (s, 6H), 3 11-3 09 (m, 4H) LC-MS(ES, m/z) 596 [(M+2H)/2]+
Example 199
Nl,N4-bis(2-(2-(2-(2-(4-(6,8-dichloro-2-methyI-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)succinamide
Figure imgf000327_0001
Compound 199, Nl,N4-bis(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinoUn-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)- ethyl)succinamide: To a solution of N-(2-(2-(2-(2 ammoethoxy)ethoxy)ethoxy)ethyl)- 4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolm-4 yl)benzenesulfonamide (compound 82) (200 mg, 0 37 mmol) in dry DMF (10 mL) under N2 was added bis(2,5- dioxopyrrolidm-1-yl) succinate (intermediate 177 1) (57 1 mg, 0 18 mmol) and tπethylamine (111 mg, 1 10 mmol) The resulting solution was stirred for 4 h at 250C in an oil bath and monitored by LCMS The resulting mixture was concentrated under vacuum and the crude product was purified by Prep-HPLC with acetonitnle water (0 05% CF3COOH)(28%-35%) This resulted m 113 8 mg (45%) of the title compound as a TFA salt 1H-NMR (300MHz, CD3OD, ppm) 7 93-7 91 (d, J-8 IHz, 4H), 7 58- 7 57 (m, 2H), 7 50-748(m, 4H), 6 87 (s, 2H), 4 88-474 (m, 4H), 4 55-4 49 (d, ./=16 2Hz, 2H), 3 94-3 88 (m, 2H), 3 67-3 59 (m, 14H), 3 55-3 45 (m, 12H), 3 35-3 09 (m, 10H), 2 48 (s, 4H) LC-MS (ES, m/z) 588 [(M+2H)/2]+
Example 200
Nl,N4-bis(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin- 4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)succinamide bis-hydrochloride salt
Figure imgf000328_0001
Intermediate 200.1, (S or R)-N-(2-(2-(2-aminoethoxy)ethoxy)ethyl)-4-(6,8-dichloro- 2-methyI-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesuIfonamlde: Intermediate 175 1 (3 g) was purified by Prep-SFC with the following conditions Column, Chiralpak IA, 2*25cm, 5um, mobile phase, CO2 (50%), iso-propanol (50%), Detector, UV 254nm This resulted in 1 g of (S or R)-N-(2-(2-(2-ammoethoxy)ethoxy)ethyl)-4- (6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinohn-4-yl)benzenesulfonamide (intermediate 200 1) as a yellow solid
Figure imgf000328_0002
Compound 200, Nl,N4-bis(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyI-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)succinamide bis-hydrochloride salt: To Intermediate 200 1 (280 mg, 0 56 mmol, 2 00 equiv) m DMF (10 mL) was added intermediate 177 1 (87 mg, 0 28 mmol, 1 00 equiv) and tnethylamme (94 3 mg, 093 mmol, 4 00 equiv) and the reaction was stirred overnight The resulting mixture was concentrated under vacuum and the crude product (300 mg) was punfied by Prep-HPLC with CH3CN H2 O (35-55%) The product was then dissolved in 15 mL of dichloromethane and gaseous hydrochloric acid was introduced for 20 minutes, then the mixture was concentrated under vacuum The crude product was washed with 3x10 mL of ether to afford 222 4 mg of Compound 200 as a light yellow solid 1H-NMR (400 MHz, CD3OD, ppm) 794-7 92 (d, J= 8Hz, 4H), 7 56-7 52 (m, 6H), 6 82 (s, 2H), 4 89-4 84 (m, 4H), 4 52-4 48 (d, J=I 64Hz, 2H), 3 91-3 90 (d, J=4Hz, 2H), 3 62-3 48 (m, 18H), 3 39-3 32 (m, 4H), 3 19-3 10 (m, 10H), 2 57-2 55 (d, J= 5 2Hz, 4H) LCMS (ES, m/z) 544 [M-2HCl]/2+H+
Example 201 2,2'-oxybis(N-(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyI-l,2,3,4- tetrahydroisoqulnolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)acetamide) bis-hydrochloride salt
Figure imgf000329_0001
Compound 201, 2,2'-oxybis(N-(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)acetamide) bis-hydrochloride salt: To intermediate 200 1 (500 mg, 1 00 mmol, 1 00 eqmv) in DMF (3 mL) was added intermediate 178 1 (150 mg, 046 mmol, 045 eqmv) and tπethylamine (0 4 g, 4 50 equiv) and the resulting solution was stirred for 2 h The crude product was purified by Prep-HPLC with CH3CN/H2O (0 05% TFA) (28%- 34%) The product was dissolved in 15 mL of dichloromethane and then gaseous hydrochloπc acid was introduced for 20 rains The mixture was concentrated under vacuum and the crude product was washed with 3x10 mL of ether to afford 101 1 mg (18%) of Compound 201 as a white solid 1H-NMR (400MHz, CD3OD, ppm) 7 94- 7 92 (m, 4H), 7 57-7 51 (m, 6H), 6 84 (s, 2H), 4 88-4 70 (m, 4H), 4 50 (s, 2H), 4 08 (s, 4H), 3 92-3 91 (m, 2H), 3 90-3 54 (m, 9H), 3 50-3 49 (m, 5H), 3 47-3 44 (m, 8H) , 3 18 (s, 6H), 3 12-3 10 (m, 4H) LCMS (ES, m/z) 552 [M-2HCl]/2+H+
Example 202 (S or R )-N,N'-(10,17-dioxo-3,6,21,24-tetraoxa-9,ll,16,18-tetraazahexacosane-l,26- diyl)bis(3-((S)-6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)benzenesulfonamide) bis-hydrochloride salt
Figure imgf000330_0001
Intermediate 202.1, (S or R)-N-(2-(2-(2-aminoethoxy)ethoxy)ethyl)-3-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide bis(2,2,2-trifluoroacetate): To 2-(2-(2-aminoethoxy)ethoxy)ethanamine (30 4 g, 205 41 mmol, 8 01 equiv) in dichloromethane (1000 mL) was added tnethylamine (5 2 g, 51 49 mmol, 2 01 equiv) This was followed by the addition of (S)-3-(6,8-dichloro-2- methyl l,2,3,4-tetrahydroisoquinolm-4-yl)benzene-l-sulfonyl chloπde hydrochloπde (10 g, 23 42 mmol, 1 00 equiv, prepared from intermediate 244 1 and the procedures descnbed in Example 1) m portions at 1O0C in 1 h The resulting solution was stirred for 15 mm at room temperature The resulting mixture was washed with 3x500 mL of bπne, dried over anhydrous sodium sulfate and concentrated under vacuum The residue was purified by Flash-Prep-HPLC with the following conditions Column, Cl 8 sihca gel, mobile phase, methanol/water/TFA (4/100/0 0005) increasing to 8/10/0 0005 within 30 mm, Detector, UV 254 nm This resulted in 7 2 g (42%) of intermediate 202 1 as a white solid
Figure imgf000331_0001
Compound 202, (S or R)-N,N'-(10,17-dioxo-3,6,21,24-tetraoxa-9,ll,16,18- tetraazahexacosane-l,26-diyl)bis(3-((S)-6,8-dichloro-2-methy]-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide) bis-hydrochloride salt: To intermediate 202 1 (500 mg, 0 69 mmol, 1 00 equiv) in DCM (10 mL) was added tπethylamine (138 mg, 1 37 mmol, 1 99 eqmv) followed by the addition of 1,4- dnsocyanatobutane (48 mg, 0 34 mmol, 0 50 equiv) in portions The resulting solution was stirred for 10 mm at room temperature then the crude product (500 mg) was puπfied by Flash-Prep-HPLC with the following conditions Column, Cl 8 silica gel, mobile phase, methanol/water=0 05/100 increasing to 90/100 within 30 min, Detector, UV 254 nm To the product was added 0 2 mL of hydrochloπc acid (2 N) and the solution lyophihzed to afford 246 7 mg (59%) of Compound 202 as a white solid 1H- NMR (400MHz, CD3OD, ppm) 7 92 (d, J=7 2Hz, 2H), 7 83 (s, 2H), 7 69-7 65 (m, 2H), 7 60-7 55 (m, 4H), 6 81 (s, 2H), 4 87-4 83 (m, 4H), 4 54-4 50 (m, 2H), 3 94-3 91 (m, 2H), 3 69-3 49 (m, 18H), 3 39-3 32 (m, 4H), 3 21-3 15 (m, 10H), 3 08-3 05 (m, 4H), 1 57 (s, 4H) LCMS (ES, m/z) 1145 [M-2H CRl]+
Example 203 (S or R)-N,N'-(2,2'-(2,2'-(2,2'-(l,4- phenylenebis(azanediyl))bis(oxomethylene)bis(azanediyl)bis(ethane-2,l- diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(3-((S or R)-6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide) bis- hydrochloride salt
HCI
Figure imgf000332_0001
Figure imgf000332_0002
Compound 203, (S or R)-N,N'-(2,2'-(2,2'-(2,2'-(l,4-phenylenebis(azanediyl))bis- S (oxomethylene)bis(azanediyl)bis(ethane-2,l-diy]))bis(oxy)bis(ethane-2,l- diyl))bis(oxy)bis(ethane-2,l-diyl))bis(3-((S or R)-6,8-dichIoro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide) bis-hydrochloride salt: To intermediate 202 1 (400 mg, 0 55 mmol, 1 00 eqmv) in DCM (10 mL) was added tπethylamme (111 mg, 1 10 mmol, 2 00 equiv) followed by the portionwise addition of
10 1,4-diisocyanatobenzene (44 mg, 028 mmol, 0 50 equiv) The resulting solution was stirred for 10 mm and the crude product (400 mg) was purified by Flash-Prep-HPLC with the following conditions Column, Cl 8 silica gel, mobile phase, methanol/water (0 05/100) increasing to 90/100 withm 30 mm, Detector, UV 254 ran To the product was added 02 mL of hydrochloric acid (2 N) and the solution lyophihzed to afford
15 201 7 mg (59%) of Compound 203 as a white solid 1H-NMR (400MHz, CD3OD, ppm) 7 84 (d, J=7 6Hz, 2H), 7 71 (s, 2H), 7 60-7 56 (m, 2H), 7 48-7 45 (m, 4H), 7 16 (s, 4H), 6 76 (s, 2H), 4 70-4 66 (m, 4H), 4 42-4 38 (m, 2H), 3 78-3 74 (m, 2H), 3 53- 3 48 (m, 18H), 3 44-3 26 (m, 4H), 3 06-2 99 (m, 10H) LCMS (ES, m/z) 1163[M- 2HC1+1]+
20
Example 204
N,N'-(butane-l,4-diyl)bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)acetamido)acetamido)acetamide)
Figure imgf000333_0001
Intermediate 204.1, 2-(2-(2-(4-(6,8-dichloro-2-methyI-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)acetamido)acetamido)acetic acid:
To a slurry of 4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqmnolin-4-yl)benzene-l- sulfonyl chloride hydrochloride (Intermediate 1.6) (283 mg, 0.66 mmol) and triglycine (152 mg, 0.80 mmol) in THF (1.5 mL) at 0 0C was added water (1.0 mL) followed by triethylamine (202 mg, 2.0 mmol). The reaction was allowed to warm to room temperature and stirred for 15 hours. The solvents were removed at reduced pressure and the residue was purified by preparative HPLC to give Intermediate 204.1 (122 mg) as a TFA salt.
Figure imgf000333_0002
Compound 204, N,N'-(butane-l,4-diyl)bis(2-(2-(2-(4-(6,8-dichloro-2-methyl- l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)acetamido)acetamido)acetamide): Intermediate 204.1 (60 mg, 0.091 πrmol) was dissoled in DMF (0.90 mL) followed by N-hydroxysuccinimide (12.6 mg, 0.11 mmol) and 1,4-diaminobutane (4.0 mg, 0.045 mmol). N-(3- dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride (21 mg, 0.11 mmol) was added and the reaction was stirred at room temperature for 16 hours, at which time additional 1,4-diaminobutane (1 uL) and N-(3-dimethylaminopropyl)-N'- ethylcarbodiimide hydrochloride (5 mg) were added. Two hours after the addition, solvent was removed at reduced pressure and the residue was purified by preparative HPLC. The title compound was obtained as a TFA salt (26 mg). 1H-NMR (400 mHz, CD3OD) δ 7.90 (d, j-8.6 Hz, 4H), 7.52 (d, j=1.8 Hz, 2 H), 7.47 (d, j=8.6 Hz, 4H), 6.84 (s, 2H), 7.75 (m, 6H), 4.44 (d, J=15.6 Hz, 2H), 3.86 (s, 4H), 3.81 (s, 4H), 3.61 (s, 4H), 3.54 (m, 2H), 3.16 (m, 4H), 3.16 (s, 6H), 1.49 (m, 4H). MS (m/z): 1636.98 [M+H]+.
Example 205
Nl,N4-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide
Figure imgf000334_0001
Compound 205, Nl,N4-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinoIta-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide: To a solution of N-(2-(2-(2-aminoethoxy)ethoxy)ethyl)-4- (6, 8-dichloro-2 -methyl- 1,2,3 ,4-tetrahydroisoquinolm-4-yl)benzenesulfonaimde (intermediate 175 1) (110 mg, 0 22 mmol) in DMF (2 0 niL) was added bis(2,5- dioxopyrrohdm-1-yl) 2,3-dihydroxysuccmate (Intermediate 168 1) (34 mg, 0 10 mmol) and the reaction was stirred for 10 minutes The solvent was removed under vacuum and the residue was purified by preparative HPLC to give the title compound (23 mg) as a TFA salt 1H-NMR (400 mHz, CD3OD) δ 7 81 (m, 4H), 7 44 (s, IH), 7 37 (m, 2H), 6 75 (s, IH), 4 64 (m, 4H), 4 37 (m, 4H), 3 72 (m, 2H), 3 46 (m, 10H), 3 38 (m, 12H), 3 02 (m, 10H) MS (m/z) 1117 02 [M+H]+
Example 206
N,N'-(2,2'-(2,2'-(2,2'-(l,4-phenylenebis(methylene))bis(azanediyl)bis(ethane-2,l- diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(3-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide)
OHC. H2N-~-^O^-O^-~-NH2 + I
Figure imgf000335_0001
Intermediate 206.1, N,N'-(l,4-phenylenebis(methylene))bis(2-(2-(2- aminoethoxy)ethoxy)ethanamine): A solution of terephthalaldehyde (134 mg, 1 0 mmol) and 2,2'-(ethane-l ,2-diylbis(oxy))diethanamine (1 48 g, 10 0 mmol) in DCM (10 mL) was stirred at room temperature After 15 minutes sodium tnacetoxyborohydπde (636 mg, 3 0 mmol) was added and the reaction was stirred for 1 5 hours Acetic acid (600 mg, 10 mmol) was then added After stirring for an additional 1 5 hours, acetic acid (600 mg, 10 mmol) and sodium tπacetoxyborohydnde (636 mg, 3 0 mmol) were added, and stirring was continued at room temperature One hour later an additional portion of sodium triacetoxyborohydride (636 mg, 3.0 mmol) was added. Twenty hours later the reaction was quenched with IN HCl (5 mL) and concentrated to dryness. Methanol (10 mL) and 12N HCl (3 drops) were added and the mixture was concentrated to dryness. The residue was dissolved in water (10 mL) and a portion (1.0 mL) was purified by preparative HPLC to give a TFA salt of the title compound (25 mg) as a TFA salt.
Figure imgf000336_0001
Compound 206, N,N'-(2,2'-(2,2'-(2,2'-(l,4- phenylenebis(methylene))bis(azanediyl)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l- diyl))bis(oxy)bis(ethane-2,l-diyl))bis(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide): To a solution of a TFA salt of intermediate 206.1 (25 mg, 0.029 mmol) in DCM (0.5 mL) was added of 4-(6,8- dichloro-2 -methyl- 1 ,2,3 ,4-tetrahydroisoquinolin-4-yl)benzene- 1 -sulfonyl chloride
(intermediate 1.6) (25 mg, 0.06 mmol) followed by triethylamine (24.2 mg, 0.24 mmol) and the reaction was allowed to stir at room temperature for 18 hours. The reaction was concentrated to dryness, and then purified by preparative HPLC to give the title compound (8 mg) as a TFA salt. 1H-NMR (400 mHz, CD3OD) δ 7.85 (m, 2H), 7.74 (m, 2H), 7.62 (m, 6H), 7.53 (m, 4H), 6.80 (s, IH), 4.74 (m, 6H), 4.44 (m, 2H), 4.30 (s, 4H), 3.83 (m, 2H), 3.76 (m, 4H), 3.62 (m, 8H), 3.50 (m, 4H), 3.23 (m, 4H), 3.10 (s, 6H), 3.02 (m, 4H). MS (m/z): 1105.05 [M+H]+.
Example 207 (2R,3R)-Nl,N4-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin- 4-yl)phenylsulfonaιnido)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccin amide
Figure imgf000337_0001
Compound 207, (2R,3R)-Nl,N4-bis(2-(2-(2-(4-(6,8-dichIoro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-2,3- 5 dihydroxysuccinamide: Following the procedures outlined in example 205, compound 207 was prepared using (2R,3R)-bis(2,5-dioxopyrrohdin-l-yl) 2,3- dihydroxysuccmate Puπfication by preparative HPLC gave a TFA salt of the title compound 1H-NMR (400 mHz, CD3OD) δ 7 82 (m, 4H), 7 45 (m, IH), 7 38 (m, 2H), 6 75 (s, IH), 4 64 (m, 4H), 4 37 (m, 4H), 3 74 (m, 2H), 3 46 (m, 10H), 3 38 (m, 12H), 10 3 02 (m, 10H) MS (m/z) 1117 07 [M+H]+
Example 208 N,N'-(13,20-dioxo-3,6,9,24,27,30-hexaoxa-12,14,19,21-tetraazadotriacontane-l,32- diyl)bis(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- I5 yl)benzenesulfonamide)
Figure imgf000337_0002
Compound 208, N,N'-(13,20-dioxo-3,6,9,24,27,30-hexaoxa-12,14,19,21-
20 tetraazadotriacontane-l,32-diyl)bis(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide): To a solution of a TFA salt of N- (2-(2-(2-(2-ammoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolm-4-yl)benzenesulfonamide (compound 28) (47 mg, 0.061 mmol) m DMF (0.20 mL) was added 1,4-dπsocyanatobutane (4.0 mg, 0 03 mmol) followed by diisopropylethylamme (15 mg, 0 12 mmol). After stirring at room temperature for 30 5 minutes, the reaction was purified by preparative HPLC to give the title compound (31 mg) as a TFA salt 1H-NMR (400 mHz, CD3OD) δ 7.88 (m, 2H), 7 75 (m, 2H), 7.63 (m, 2H), 7.54 (m, 4H), 6.83 (m, 2H), 474 (m, 4H), 4.48 (m, 2H), 3.87 (m, 2H), 3 62- 3.55 (m, 14H), 3.51-3.43 (m, 12H), 3.24 (m, 4H), 3 14 (s, 6H), 3 05 (m, 8H), 1 43 (m, 4H). MS (m/z): 1230.99 [M+H]+. 10
Example 209
N,N'-(l,l'-(l,4-phenylenebis(azanediyl))bis(l-oxo-5,8,ll-trioxa-2-azatridecane- 13,l-diyl))bis(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)benzenesulfonamide) I5
Figure imgf000338_0001
Compound 209, N,N'-(l,l'-(l,4-phenylenebis(azanediyl))bis(l-oxo-5,8,ll-trioxa-2- azatridecane-13,l-diyl))bis(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin- 20 4-yl)benzenesulfonamide): Following the procedures outlined in example 208, compound 209 was prepared using 1 ,4-diisocyanatobenzene. Purification by preparative HPLC gave a TFA salt of the title compound 1H-NMR (400 mHz, CD3OD) δ 7 78 (m, 2H), 7.64 (m, 2H), 7 53 (m, 2H), 7 43 (m, 2H), 7.39 (m, 2H), 7.10 (s, 4H), 6.71 (s, 2H), 4 58 (m, 4H), 4 39 (m, 2H), 3 68 (m, 2H), 3 54 (s, 8H), 3 50 - 3 44 (m, 8H), 3.42 (m, 6H), 3.35 (m, 4H), 2.99 (s, 6H), 2.95 (m, 4H). MS (m/z): 1250.98 [M+H]+.
Example 210 (2R,3R)-Nl,N4-bis(20-(4-(4-((E)-3-(diaminomethyleneamino)-2-methyl-3-oxoprop- l-enyl)-2,6-difluorophenoxy)phenyIsulfonamido)-3,6,9,12,15,18-hexaoxaicosyI)-2,3- dihydroxysuccinamide
Figure imgf000339_0001
Intermediate 210.1, (E)-ethyl 3-(4-(4-(N-(20-amino-3,6,9,12,15,18- hexaoxaicosyl)sulfamoyl)phenoxy)-3,5-difluorophenyl)-2-methylacrylate:
Intermediate 210.1 was prepared following the procedure outlined in Example 44.2 using 20-azido-3,6,9, 12,15, 18-hexaoxaicosan-l-amine. The title compound was recovered in 64% yield as a yellow oil.
Figure imgf000339_0002
OZTLP O O O U ™ ! O
Intermediate 210.2, (2R,3R)-Nl,N4-bis(20-(4-(4-((E)-4-(2-carboxyprop-l-enyl)-2,6- difluorophenoxy)phenyIsulfonamido)-3,6,9,12,15,18-hexaoxaicosy])-2,3- dihydroxysuccinamide. Intermediate 210.2 was prepared following the procedure outlined in Example 168 using (2R,3R)-bis(2,5-dioxopyrrolidin-l-yl) 2,3- dihydroxysuccinate (22.4mg, 0.065mmol) and (E)-ethyl 3-(4-(4-(N-(20-amino- 3,6,9,12,15,18-hexaoxaicosyl)sulfamoyl)phenoxy)-3,5-difluorophenyl)-2- methylacrylate (91.5mg, 0.13mmol). The title compound was recovered in 60% yield as a clear semi-solid.
Figure imgf000340_0001
Compound 210, (2R,3R)-Nl,N4-bis(20-(4-(4-((E)-3-(diaminomethyleneamino)-2- methyl-3-oxoprop-l-enyl)-2,6-difluorophenoxy)phenylsulfonamido)-3,6,9, 12, 15,18- hexaoxaicosyI)-2,3-dihydroxysuccinamide. Compound 210 was prepared following the procedure outlined in Example 45 using Intermediate 210.2 (59.6mg). Purification 10 by preparative HPLC gave the title compound (lOmg) as a TFA salt. 1H-NMR (400MHz, CD3OD): 57.64 (d, 4H), 7.48 (s, IH), 7.32 (d, 4H), 7.12 (d, 4H), 3.62-3.58 (m, 17H), 3.55-3.52 (m, 9H), 3.48-3.41 (m, 13H), 3.06 (s, 3H), 2.72 (s, 6H) . MS (ra/z): 1549.23 [M+H]+.
I5 Compound 211
(E)-3-(4-(4-(N-(20-amino-3,6,9,12,15,18-hexaoxaicosyl)sulfamoyl)phenoxy)-3,5- difluorophenyl)-N-(diaminomethylene)-2-methyIacry]amide
Figure imgf000340_0002
20
Compound 211, (E)-3-(4-(4-(N-(20-amino-3,6,9,12,15,18- hexaoxaicosyl)sulfamoyl)phenoxy)-3,5-difluorophenyl)-N-(diaminomethy]ene)-2- methylacrylamide: Compound 211 was prepared following the procedure outlined in Example 45 using (E)-ethyl 3-(4-(4-(N-(20-amino-3,6,9,12,15,18-
25 hexaoxaicosyl)sulfamoyl)phenoxy)-3,5-difluorophenyl)-2-methylacrylate (Intermediate
210.2, 13.2mg). Purification by preparative HPLC gave the title compound (8.7mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 84 (d, 2H), 7 52 (s, IH), 7 35 (d, 2H), 7 12 (d, 2H), 3 74-3 70 (m, 2H), 3 69-3 58 (m, 24H), 3 55-3 51 (m, 2H), 3 49-3 46 (m, 2H), 3 15-3 12 (m, 2H), 3 07-3 04 (m, 2H) MS (m/z) 718 28 [M+H]+
Example 212
(2R,3R)-Nl,N4-bis(2-(2-(2-(2-(4-(4-((E)-3-(diaminomethyleneamino)-2-methyl-3- oxoprop-l-enyI)-2,6- difluorophenoxy)phenyIsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide
Figure imgf000341_0001
Intermediate 212.1, (E)-ethyl 3-(4-(4-(N-(2-(2-(2-(2- aminoethoxy)ethoxy)ethoxy)ethyl)sulfamoyl)phenoxy)-3,5-difluorophenyl)-2- methylacrylate: Compound 44 2 (lOOmg, 0 175mmol) and (2R,3R)-bis(2,5- dioxopyrrohdin-1-yl) 2,3-dihydroxysuccmate (30 lmg, 0087mmol) were dissolved in DMF (0 35mL) with DIEA (67 7mg, 0 525mmol) and stiired for 2 hours at room temperature The solvent was removed and the resulting material partitioned between EtOAc (2OmL) and water (2OmL) The organic layer was washed with saturated NaHCO3 (2OmL), bπne (2OmL) and dried over Na2SO4 to give the product (87 7mg) as a yellow oil that was used without further purification
Figure imgf000342_0001
Compound 212, (2R,3R)-Nl,N4-bis(2-(2-(2-(2-(4-(4-((E)-3-
(diaminomethyleneamino)-2-methyI-3-oxoprop-l-enyl)-2,6- difluorophenoxy)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide: Compound 212 was prepared following the procedures outlined in Example 45 . Purification by preparative HPLC gave 9.6mg of the title compound as the TFA salt. 1H-NMR (400MHz, CD3OD): δ 7.86 (d, 4H), 7.44 (s, 2H), 7.31 (d, 4H), 7.11 (d, 4H), 4.44 (s, 2H), 3.61-3.53 (m, 21H), 3.50-3.41 (m, 15H), 3.05 (t, 4H), 2.17 (s, 6H). MS (m/z): 1286.11 [M+H]+.
Example 213 2,2',2"-nltrilotris(N-(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)acetamide)
Figure imgf000343_0001
Compound 213, 2,2',2"-nitrUotris(N-(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)- ethy])acetamide): Compound 213 was prepared following the procedure outlined in Example 168 using tπs(2,5-dioxopyrrohdin-l-yl) 2,2',2"-mtπlotπacetate (75mg, 0 156mmol) and N (2-(2 (2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoqumohn-4-yl)benzenesulfonamide (Compound 28, 254mg, 0467mmol) Purification by preparative HPLC gave the title compound (32 Omg) as the TFA salt 1H-NMR (400MHz, CD3OD) δ 7 88 (d, 3H), 7 75 (s, 3H), 7 63 (t, 3H), 7 54 (t, 6H), 6 82 (s, 3H), 4 84-4 75 (m, 6H), 448 (d, 3H), 3 86 (m, 3H), 3 85-3 37 (m, 54H), 3 14 (s, 9H), 3 02 (t, 6H) MS (m/z) 1777 07 [M+H]+
Example 214 N-(32-amino-3,6,9,12,15,18,21,24,27,30-decaoxadotriacontyl)-3-(6,8-dichIoro-2- methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide
Figure imgf000344_0001
Intermediate 214.1, N-(32-azido-3,6,9,12,15,18,21,24,27,30-decaoxadotriacontyl)-3- (6,8-dichloro-2-methyI-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide: A solution of 32-azido-3,6,9,12,15,18,21,24,27,30-decaoxadotnacontan-l-amine (436 9mg, 0 777mmol) in dry DMF (3 5mL) under N2 was cooled to 00C A solution of 3-(6,8-dichloro-2-methyl-l, 2,3,4 tetrahydroisoqumolm 4-yl)benzene-l-sulfonyl chlonde (300mg, 0 706mmol) and DIEA (273 2mg, 2 118mmol) in DMF (3mL) was added dropwise After 60 minutes LCMS indicated complete conversion and the solvent was removed to give N-(32-azido-3,6,9,12,15,18,21,24,27,30- decaoxadotnacontyl)-3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolm-4- yl)benzenesulfonamide (620mg) as a yellow oil which was used without further purification
Figure imgf000344_0002
Compound 214, N-(32-amino-3,6,9,12,15,18,21,24,27,30-decaoxadotriacontyl)-3- (6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide: To a solution of N-(32-azido-3,6,9,12,15,18,21,24,27,30-decaoxadotπacontyl)-3-(6,8- dichloro-2-methyl- 1 ,2,3 ,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide
(Intermediate 214 1, 620mg, 0 706mmol) in THF/H2O (10 1 v/v, 14 3mL) under N2 was added tπmethylphosphme (214 8mg, 2 82mmol) The resulting solution was stirred overnight at which point LCMS indicated complete conversion The solvent was removed to give 819 mg of an orange oil, a portion of which was purified by preparative HPLC to give the title compound as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 90 (d, IH), 7 68 (s, IH), 7 62 (t, IH), 7 55 (m, 2H), 6 82 (s, IH), 3 85 (m, IH), 3 78 (q, 3H), 3 70-3 58 (m, 55H), 3 52 (m, 2H), 3 46 (t, 3H), 3 18 (t, 3H), 3 11 (s, 3H), 3 03 (t, 2H) MS (m/z) 855 24 [M+H]+
Example 215
Nl,N3,N5-tris(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethy])benzene-l,3,5-tricarboxamide
Figure imgf000345_0001
Compound 215, Nl,N3,N5-tris(2-(2-(2-(2-(3-(6,8-diehloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4- yl)phenylsulfonamldo)ethoxy)ethoxy)ethoxy)ethyl)benzene-l,3,5-tricarboxamide:
To a solution of N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8 dichloro-2- methyl-l,2,3,4-tetrahydroisoqmnolin-4-yl)benzenesulfonamide (Compound 28, 75mg, 0 0968) in DMF (0 5mL) was added benzene-l,3,5-tπcarboxyhc acid (6 7mg, 0 0319mmol), DIEA (37 5mg, 0 291mmol), and finally HATU (40 4mg, 0 107mmol) The reaction was stirred for 60 mmutes at room temperature at which point LCMS indicated complete conversion The resulting solution was diluted with acetomtπle/water solution (1 1 v/v) and filtered Purification by preparative HPLC gave the title compound (37 7mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 8 37 (s, 3H), 7 84 (d, 2H(, 7 83 (s, 2H), 7 62 (t, 2H), 7 51-7 50 (m, 4H), 6 79 (s, 2H), 4 83- 4 70 (m, 5H), 4 46 (d, 2H), 3 86 (q, 2H), 3 67-3 53 (m, 27H), 3 45 (t, 5H), 3 39 (t, 5H), 3 14 (s, 7H), 2 98 (t, 4H) MS (m/z) 1797 15 [M+H]+
Example 216
Nl,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)terephthalamide
Figure imgf000346_0001
Compound 216, Nl,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)pheny]sulfonamido)ethoxy)ethoxy)ethoxy)- ethyOterephthalamide: Compound 216 was prepared following the procedure outlined in Example 215 using terephthalic acid (10 7mg, 0 0646mmol) and N-(2-(2-(2- (2-ammoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro 2 methyl- 1,2,3, 4- tetrahydroisoqumolm-4-yl)benzenesulfonamide (Comopound 28, lOOmg, 0 129mmol) Purification by preparative HPLC gave the title compound (46 3mg) as a TFA salt 1H NMR (400MHz, CD3OD) δ 7 87 (m, 6H), 7 73 (s, 2H), 7 59 (t, 2H), 7 52-7 49 (m, 4H)m, 6 80 (s, 2H), 4 77-4 69 (m, 4H), 449 (d, 2H), 3 587 (qs, 2H), 3 67 3 54 (m, 27H), 3 45 (t, 5H), 3 40 (t, 5H), 3 13 (s, 7H), 2 99 (t, 4H) MS (m/z) 1224 34 [M+H]+
Example 217 Nl,N31-bis(32-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)-3,6,9,12,15,18,21,24,27,30-decaoxadotriacontyl)-
4,7,10,13,16,19,22,25,28-nonaoxahentriacontane-l,31-diamide
Figure imgf000347_0001
Compound 217, Nl,N31-bis(32-(3-(6,8-dichloro-2-methyL-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)-3,6,9,12,15,18,21,24,27,30- decaoxadotriacontyl)-4,7,10,13,16,19,22,25,28-nonaoxahentriacontane-l,31- diamide: Compound 217 was prepared following the procedure outlined m Example 168 using bis(2,5-dioxopyrrolidin-l-yl) 4,7,10,13,16,19,22,25,28- nonaoxahentnacontane-l ,31-dioate (69 lmg, 0 0975mmol) and N-(32-ammo- 3,6,9,12,15,18,21,24,27,30-decaoxadotπacontyl)-3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoqumolm-4-yl)benzenesulfonamide (Compound 214, 166 2mg, 0 195mmol) Puπfication by preparative HPLC gave the title compound (106 3mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 88 (d, 2H), 7 76 (s, 2H), 7 66 (t, 2H), 7 56 (m, 4H), 6 86 (s, 2H), 3 90 (m, 2H), 3 82 (t, 2H), 3 76 (m, 6H), 3 62-3 41 (m, 28H), 3 38 (m, 6H), 3 35-3 28 (m, 56H), 3 15 (s, 6H), 3 05 (t, 4H), 2 43 (t, 4H) MS (m/z) 1094 37 [(M+2H)/2]+
Example 218
2R,3R)-Nl,N4-bis(2-(2-(2-(3-(6,8-dichIoro-2-methyl-l,2,3,4-tetrahydroisoquinoIin- 4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide:
Figure imgf000348_0001
Compound 218, (2R,3R)-Nl,N4-bis(2-(2-(2-(3-(6,8-dichloro-2-methyI-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide: Compound 218 was prepared following the procedure outlined in Example 168 using (2R,3R)-bis(2,5-dioxopyrrolidm-l-yl) 2,3- dihydroxysuccmate (102mg, 00298mmol) and N-(2-(2-(2-ammoethoxy)ethoxy)ethyl)- 3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolin-4-yl)benzenesulfonamide (Compound 168 2, 30mg, 0 0597mmol) Purification by preparative HPLC gave the title compound (5 lmg) as the TFA salt 1H-NMR (400MHz, CD3OD) δ 7 92 (d, J=I 8Hz, 2H), 7 82(m, 2H), 7 67 (t, 7=7 8Hz, 2H), 7 57(m, 2H), 7 55 (d, J=6 9Hz, 2HO, 6.86 (m, 2H), 4 84(s, 2H), 4 79(s, 2H) , 4 54(d, 2H), 448(s, 2H), 3 92(m, 2H) , 3 53(m, 22H) , 3 18(s, 6H), 3 07(t, J=S 4Hz, 4H) MS (m/z) 1119 04 [M+H]+
Example 219
Nl,N3-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- y])phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)benzene-l,3-disulfonamide
Figure imgf000348_0002
Compound 219, Nl,N3-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)benzene-l,3-disulfonamide: To a solution of N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoqmnolin-4-yl)benzenesulfonamide (Compound 28, 50mg, 0 0917mmol) and DIEA (35 5mg, 0275mmol) in dry DCM (0 183mL) under N2 was added benzene-l,3-disulfonyl dichloπde (12 7mg, 00459mmol) in DCM (0 183mL) The reaction mixture was stirred at room temperature for 60 minutes at which point LCMS indicated complete conversion The solvent was removed and the resulting
10 residue brought up m 4 mL ACN/H2O solution (1 1) Filtration and purification by preparative HPLC gave the title compound (16 6mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 8 28 (s, IH), 8 06 (d, IH), 7 85 (d, 2H), 7 75 (d, 2H), 7 70 (s, IH), 7 63 (t, 2H), 7 53 (m, 3H), 6 82 (s, IH), 4 52 (d, IH), 3 85 (d, IH), 3 61-3 46 (m, 28H), 3 13 (s, 6H), 3 09-3 03 (m, 7H) MS (Wz) 1294 99 [M+H]+
I5
Example 220
N4,N4'-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)biphenyl-4,4'-disulfonamide
Figure imgf000349_0001
Compound 220, N4,N4'-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)- ethyl)biphenyl-4,4'-disulfonamide: Compound 220 was prepared following the
25 procedure outlined in Example 219 using biphenyl-4,4'-disulfonyl dichloπde (16 lmg, 0 0459mmol) and N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl) 3-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoquinolm-4-yl)benzenesulfonamide (Compound 28, 50mg, 0 0917mmol) Punfication by preparative HPLC gave the title compound (16 7mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 96 (d, 4H), 7 88-7 85 (m, 5H), 7 78 (s, 2H), 7 61 (t, 2H), 747 (d, 2H), 678 (s, 2H), 4 74-4 69 (m, 3H), 445 (d, 2H), 3 88-3 83 (m, 2H), 3 62-3 59 (m, 2H), 3 55-3 53 (m, 9H), 3 52-3 43 (m, 17H), 3 13 (s, 6H), 3 11- 3 03 (m, 8H) MS (m/z) 1371 02 [M+H]+
Example 221
(14R,15R)-l-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- 10 yl)phenylsuIfonainido)-14,15-dihydroxy-13-oxo-3,6,9-trioxa-12-azahexadecan-16- oic acid
Figure imgf000350_0001
I5 Compound 221, (14R,15R)-l-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yI)phenylsulfonamido)-14,15-dihydroxy-13-oxo-3,6,9- trioxa-12-azahexadecan-lό-oic acid: Compound 221 was prepared by isolating the mono-addition byproduct from the procedure outlined in Example 168 using (2R,3R)- bis(2,5 dioxopyrrolidin-1-yl) 2,3-dihydroxysuccinate (704mg, 0205mmol) and
20 Compound 28 (223mg, 0409mmol) Punfication by preparative HPLC gave the title compound (44 4mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 89 (d, IH), 7 81 (d, IH), 7 63 (t, IH), 7 55 (s, IH), 7 50 (t, IH), 6 84 (s, 0 5H), 3 88-3 84 (m, IH), 3 64- 3 34 (m, 22H), 3 14 (s, 4H), 3 07 (m, 2H) MS (m/z) 677 36 [M+H]+
2S Example 222 (2S,3S)-Nl,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-raethyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethy])-2,3- dihydroxysuccinamide
Figure imgf000351_0001
Compound 222, (2S,3S)-Nl,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l ,2,3,4- tetrahydroisoquinolin-4-yI)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide: Compound 222 was prepared following the procedure
10 outlined in Example 215 using (2S,3S)-2,3-dihydroxysuccimc acid (15 5mg, 0 103mmol) and N-(2-(2-(2-(2-ammoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoqumolm-4-yl)benzenesulfonamide (Compound 28, 112mg, 0206mmol) Puπfication by preparative HPLC gave the title compound (39 9mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 87 (d, 2H), 7 77 (s, 2H), 7 63 (t, 2H),
15 7 54-7 50 (m, 4H), 6 82 (s, 2H), 434 (s, 2H), 3 90-3 85 (m, IH), 3 62-3 30 (m, 47H), 3 14 (m, 8H), 3 05 (t, 4H) MS (m/z) 1206 95 [M+H]+
Example 223
Nl,N4-bis(2-(2-(2-(2-(3-((R)-6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-
2Q yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide
Figure imgf000352_0001
Intermediate 223.1a, (R or S)-N-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-3- (6,8-dichIoro-2-raethyI-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide and 223.1b (S or R)-N-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethy])-3-(6,8-dichIoro-2- methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide: N-(2-(2-(2-(2- azidoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide (intermediate 28.1, 4.5 g, 7.88 mmol, 1.00 equiv) was separated into its enantiomers by chiral phase preparative Supercritical
10 Fluid Chromatography (Prep-SFC) with the following conditions: Column, Chiralpak IA, 2*25cm, 5um; mobile phase, CO2(80%), methanol (20%); Detector, UV 254nm.
This resulted in 1.61 g of (R or S)-N-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-3- (6,8-dichloro-2-methyl- 1 ,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide as a I5 yellow oil. 1H-NMR (300MHz, CD3OD, ppm): δ 7.79 (d, J=7.5Hz, IH), 7.711 (s, IH), 7.49-7.58 (m, 2H), 7.36-7.37 (m, IH), 6.83 (s, IH), 4.40-4.44 (m, IH), 3.80 (d, J=16.2Hz, IH), 3.58-3.69 (m, 9H), 3.40-3.52 (m, 4H), 3.33-3.38 (m, 3H), 3.03-3.09 (m, 3H), 2.66-2.72 (m, IH), 2.50 (s, 3H). MS (m/z): 572 [M+H]+.
20 This also gave 1.81 g of (S or R)-N-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-3- (6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide as yellow oil 1H-NMR (300MHz, CD3OD, pprriy δ 7 78-7 81 (m, IH), 7 71 (s, IH), 749-7.58 (m, 2H), 7.36-7.37 (m, IH), 6.83 (s, IH), 4.40-4.44 (m, IH), 3.80 (d, /=15.9Hz, IH), 3.57-3.70 (m, 9H), 3.44-3 53 (m, 4H), 3.37-3.40 (m, 3H), 3.03-3.09 (m, 3H), 2.66-2.72 (m, IH), 2 50 (s, 3H). MS (m/z): 572 [M+H]+.
Figure imgf000353_0001
Intermediate 223.2, (R or S)-N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-3- (6,8-dichloro-2-methyH,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide:
10 Following the procedure outlined m example 170, intermediate 223 Ia was converted to Intermediate 223 2.
Figure imgf000353_0002
I5 Compound 223, Nl,N4-bis(2-(2-(2-(2-(3-((R or S)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide: Compound 223 was prepared following the procedures outlined in Example 168 using (R or S)-N-(2-(2-(2-(2- ammoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l, 2,3,4-
20 tetrahydroisoqumohn-4-yl)benzenesulfonamide (intermediate 223 2, 239mg, 0439mmol) and bis(2,5-dioxopyrrohdm-l-yl) 2, 3 -dihydroxy succinate (75 5mg,
0.219mmol) Purification by preparative HPLC gave the title compound (135 5mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 89 (d, 2H), 7 68 (s, 2H), 7 63 (t, 2H), 7 54-7 52 (m, 4H), 6 83 (s, 2H), 4 83-4 75 (m, 5H), 4 50-4 48 (m, 2H), 443 (d, 2H), 3 89-3 82 (m, 2H), 3 63-3 35 (m, 34H), 3 14 (s, 6H), 3 04 (t, 4H) MS (m/z) 1208 11 [M+H]+
Example 224
Nl,N4-bis(2-(2-(2-(2-(3-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinoIin-4-yl)phenylsulfonaniido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide
Figure imgf000354_0001
Intermediate 224.1, (S or R)-N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-3- (6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide: Following the procedure outlined in example 170, intermediate 223 Ib was converted to Intermediate 224 1
Figure imgf000354_0002
Compound 224, Nl,N4-bis(2-(2-(2-(2-(3-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide: Compound 224 was prepared following the procedures outlined in Example 223 using (S or R)-N-(2-(2-(2-(2- aminoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2 -methyl 1,2,3,4 tetrahydroisoquinolm-4-yl)benzenesulfonamide (intermediate 224.1, 274mg, 0 502mmol) and bis(2,5-dioxopyrrobdin-l-yl) 2,3-dihydroxysuccinate (86.4mg, 0.251mmol). Puπfication by preparative HPLC gave the title compound (159mg) as a TFA salt. 1H-NMR (400MHz, CD3OD) δ 7.87 (d, 2H), 7.77 (s, 2H), 7.63 (t, 2H), 6 54-6 51 (m, 4H), 6.83 (s, 2H), 4 84-4.75 (m, 4H), 4.50-4.43 (m, 4H), 3.90-3.85 (m, 4H), 3.62-3.28 (m, 35H), 3.14 (s, 6H), 3.04 (t, 4H). MS (m/z). 1207.11 [M+H]+.
Example 225
Nl,N4-bis(2-(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolta-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccin amide
Figure imgf000355_0001
Intermediate 225.1a, (R or S)-N-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-4- (6,8-dichloro-2-methyI-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide and intermediate 225.1b, (S or R)-N-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-4- (6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesuIfonamide:
N-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-4-(6,8-dichloro-2-methyl-l , 2,3,4- tetrahydroisoqumohn-4-yI)benzenesulfonamide (5 g, 8 76 mmol, 1 00 equiv) was separated into its enantiomers by Prep-SFC with the following conditions. Column, Chiralpak IA, 2*25cm, 5um, mobile phase, CO2 (80%), ethanol (20%), Detector, UV 254nm
This resulted m 1 69 g of (R or S)-N-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-4- (6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolm-4-yl)benzenesulfonamide as a brown oil 1H-NMR (300MHz, CD3OD, ppm) δ 7 85 (d, JS 4Hz, 2H), 7 40 (d, JS IHz, 2H), 7 36 (s, IH), 6 82 (s, IH), 443 (t, IH), 3 81 (m, IH), 3 67 (m, 9H), 3 48 (m, 4H), 3 33 (m, 2H), 3 01 (m, IH), 2 71 (m, IH), 2 49 (s, 3H) MS (m/z) 572 [M+H]+
Also isolated was 1 65 g of (S or R)-N-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-4- (6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolin-4-yl)benzenesulfonamide as brown oil 1H-NMR (300MHz, CD3OD, ppm) δ 7 84 (d, /"8 4Hz, 2H), 7 43 (d, JS IHz, 2H), 7 36 (s, IH), 6 82 (s, IH), 442 (t, IH), 3 81 (m, IH), 3 67 (m, 10H), 3 59 (m, 4H), 3 49 (m, 2H), 3 11 (m, 2H), 2 72 (m, IH), 249 (s, 3H) MS (m/z) 572 [M+H]+
Figure imgf000356_0001
Intermediate 225.2, (S or R)-N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-4- (6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesu]fonamide:
Following the procedure outlined in example 170, intermediate 225 Ib was converted to Intermediate 225 2
Figure imgf000357_0001
Compound 225, Nl,N4-bis(2-(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolm-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide: Compound 225 was prepared following the procedures outlined in Example 168 using (S)-N-(2-(2-(2-(2-ammoethoxy)ethoxy)ethoxy)ethyl)-4- (6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolm-4-yl)benzenesulfonamide (intermediate 225 2, 302 4mg, 0 555mmol) and bis(2,5-dioxopyrrolidm 1 yl) 2,3- dihydroxysuccinate (95 5mg, 0277mmol) Puπfication by preparative HPLC gave the title compound (97 lmg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 85 (d, 4H), 7 54 (s, 2H), 746 (d, 4H), 6 84 (s, 2H), 4 88-4 72 (m, 3H), 443-442 (m, 2H), 3 85-3 80 (m, IH), 3 63-3 35 (m, 24H), 3 13 (s, 5H), 3 08 (t, 4H) MS (m/z) 1208 05 [M+H]+
Example 226 Nl,N4-bis(2-(2-(2-(2-(4-((R or S)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide
Figure imgf000358_0001
Intermediate 226.1, (R or S)-N-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-4-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide: Following the procedure outlined in example 170, intermediate 225 Ia was converted to intermediate 226 1
Figure imgf000358_0002
Compound 226, Nl,N4-bis(2-(2-(2-(2-(4-((R or S)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide: Compound 226 was prepared following the procedures outlined in Example 168 using (R or S) N-(2-(2 (2 (2 aminoethoxy)ethoxy)emoxy)ethyl)-4-(6,8-dichloro-2-methyl- 1,2,3,4- tetrahydroisoquinolm-4 yl)benzenesulfonamide (intermediate 226 1, 267 5 mg, 0491mmol) and bis(2,5 dioxopyrrolidin 1 yl) 2,3 dihydroxysuccinate (84 5mg, 0 245mmol) Purification by preparative HPLC gave the title compound (145 4mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 89 (d, 5H), 7 54 (s, 2H), 7 48 (d, 4H), 6 84 (s, 2H), 4 84-473 (m, 4H), 4 50-443 (d, 2H), 4 18 (d, 2H), 3 85-3 80 (m, 2H), 3 64- 3 40 (m, 32H), 3 13 (s, 6H), 3 08 (t, 3H) MS (m/z) 1207 10 [M+H]+
Example 227
Nl,N4-bis(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinoUn-4- yl)phenylsuJfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide
Figure imgf000359_0001
Compound 227, Nl,N4-bis(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinoIm-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide: Compound 227 was prepared following the procedure outlined in Example 168 using bis(2,5-dioxopyrrohdin-l-yl) 2,3-dihydroxysuccinate (49 6mg, 0 144mmol) and N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-4-(6,8- dichloro-2-methyl- 1 ,2,3 ,4-tetrahydroisoquinolm-4-yl)benzenesulfonamide (Compound 82, 157mg, 0288mmol) Puπfication by preparative HPLC gave the title compound (34 5mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 89 (d, 4H), 7 53 (s, 2H), 7 45 (d, 4H), 6 83 (s, 2H), 4 77-4 74 (m, 6H), 4 46 (d, 2H), 443 (t, 2H), 3 89-3 84 (m, 2H), 3 62 3 53 (m, 19H), 3 49-3 41 (m, 13H), 3 14 (s, 6H), 3 08 (t, 4H) MS (m/z) 1206 94 [M+H]+
Example 228
Nl,N3-bis(2-(2-(2-(2-(3-(6,8-dichIoro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)isophthalamide
Figure imgf000360_0001
Compound 228, Nl,N3-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)- ethyl)isophthalamide: Compound 228 was prepared following the procedure outlined in Example 215 using isophthalic acid (8 0 mg, 0.0484 mmol) and N-(2-(2-(2-(2- ammoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoqumolm-4-yl)benzenesulfonamide (Compound 28, 75mg, 0 0968mmol) Puπfication by preparative HPLC gave the title compound (45 6mg) as a TFA salt. 1H- NMR (400 MHz, CD3OD)- δ 8 25 (s, IH), 7 92 (d, 2H), 7 85 (d, 2H), 7 73 (s, 2H), 7 58 (t, 2H), 7 49 (m, 5H), 6.81 (s, 2H), 4.83-4.71 (m, 4H), 4.49 (d, 2H), 3 87 (m, 2H), 3 67- 3 54 (m, 28H), 3 45 (t, 5H), 3 44 (q, 5H), 3 14 (s, 7H), 2.99 (t, 4H). MS (m/z): 1223.19 [M+H]+
Example 229
(2R,3S)-Nl,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyI-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide
Figure imgf000360_0002
Compound 229, (2R,3S)-Nl,N4-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyI)-2,3- dihydroxysuccinamide: N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide (Compound 28, 25mg, 0.0322mmol) was dissolved in DMF (0.16ImL) with DIEA (12.4mg, 0.0966mmol) and (2R,3S)-2,3-dihydroxysuccinic acid (2.7mg, 0.0161mmol). Benzotriazol-1-yl-oxytripyrrolidinophosphonium hexafluorophosphate (PyBOP) (18.4mg, 0.0354mmol) was added and the resulting solution stirred for 60 minutes, at
10 which point LCMS indicated complete conversion. The reaction mixture was diluted to 2 mL with acetonitrile/water (1 :1) and filtered. Purification by preparative HPLC gave the title compound (8.7mg) as a TFA salt. 1H-NMR (400MHz, CD3OD): δ 7.80 (d, 2H), 7.69 (s, 2H), 7.55 (t, 2H), 7.43 (m, 4H), 6.75 (s, 2H), 4.80-4.75 (m, 3H), 4.39 (d, 2H), 4.24 (d, 2H), 3.76 (m, 2H), 3.64-3.25 (m, 33H), 3.04 (s, 7H), 2.95 (t, 4H) . MS
I5 (m/z): 1207.10 [M+H]+.
Example 230
Nl,N2-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)phthalamide
Figure imgf000361_0001
Compound 230, Nl,N2-bis(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenyIsulfonamido)ethoxy)ethoxy)ethoxy)- ethyl)phthalamide: Compound 230 was prepared by following the procedure outlined in Example 215 using phthalic acid (8.0mg, 0.0484mmol) and N-(2-(2-(2-(2- aminoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide (Compound 28, 75mg, 0.0968 mmol). Purification by preparative HPLC gave the title compound (35.4mg) as a TFA salt. 1H- NMR (400MHz, CD3OD): δ 7.87 (d, 2H), 7.76 (s, 2H), 7.63 (t, 2H), 7.50 (m, 8H), 6.79 (s, 2H), 4.83-4.73 (m, 4H), 4.65 (d, 2H(), 3.85 (q, 2H), 3.62-3.39 (m, 36H), 3.10 (s, 6H), 3.02 (t, 4H) . MS (m/z): 1223.00 [M+H]+.
Example 231
Nl,N4-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)terephthalamide
Figure imgf000362_0001
Compound 231, Nl,N4-bis(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinoIin-4-yL)phenylsulfonamido)ethoxy)ethoxy)ethyl)- terephthalamide: Compound 231 was prepared following the procedure outlined in Example 215 using terephthalic acid (11.4mg, 0.0684mmol) and 4-(6,8-dichloro-2- methyl- 1 ,2,3 ,4-tetrahydroisoquinolin-4-yl)-N-(2-(2-(2-hydroxyethoxy)ethoxy)-
36Q ethyl)benzenesulfonamide (Compound 175 1, lOOmg, 0 136mmol) Purification by preparative HPLC gave the title compound (9 8mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 86-7 85 (m, 9H), 7 83 (s, 2H), 7 50 (s, IH), 741 (d, 4H), 6 80 (s, IH), 3 68-3 42 (m, 26H), 3 34 (m, 2H), 3 09-3 Ol (m, 12H) MS (m/z) 1135 07 [M+H]+
Example 232
N,N'-(10-oxo-3,6,14,17-tetraoxa-9,ll-diazanonadecane-l,19-diyl)bis(4-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide)
Figure imgf000363_0001
Compound 232, N,N'-(10-oxo-3,6,14,17-tetraoxa-9,ll-diazanonadecane-l,19- diyl)bis(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)benzenesulfonamide): N-(2-(2-(2-ammoethoxy)ethoxy)ethyl)-4-(6,8-dichloro-2- methyl- 1, 2,3 ,4-tetrahydroisoqumohn-4-yl)benzenesulfonamide (Compound 175 1, 80mg, O l lOmmol) and DIEA (42 lmg, 0 330mmol) were dissolved in dry DCM (0 5mL) under N2 and cooled to O°C A solution of tnphosgene (4 9mg, 0 0165mmol) in DCM (0 2mL) was added dropwise and the resulting solution was warmed to room temperature over 30mmutes The solvent was removed, the resulting residue was brought up in 4 mL of acetomtπle/water (1 1) solution and filtered Purification by preparative HPLC gave the title compound (8 5mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 90 (d, 4H), 7 60 (s, 2H), 747 (d, 4H), 6 84 (s, 2H), 3 58-3 42 (m, 24H), 3 12 3 05 (m, 17H) MS (m/z) 1031 96 [MH-H]+ Example 233
NlrN4-bis(2-(2-(2-(2-(4-(6,8-dlchloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)terephthalamide
Figure imgf000364_0001
Compound 233, Nl,N4-bis(2-(2-(2-(2-(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenyIsulfonamido)ethoxy)ethoxy)ethoxy)- ethyl)terephthalamide: Compound 233 was prepared following the procedures
10 outlined in Example 215 using terephthalic acid (10 4mg, 0 0628mmol) and N-(2-(2-(2- (2-ammoethoxy)ethoxy)ethoxy)emyl)-4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoqumolm-4-yl)benzenesulfonamide (Compound 82, 97 2mg, 0 1255mmol) Punfication by preparative HPLC gave the title compound (38.9mg) as a TFA salt. 1H- NMR (400MHz, CD3OD) δ 7 83 (m, 10H), 7.85 (s, 2H), 7 42 (d, 4H), 6 83 (s, IH),
15 3 66-3 55 (m, 28H), 3 46-3 39 (m, 1 IH), 3 12 (s, 7H), 3 04 (t, 4H) MS (m/z) 1223 14 [M+H]+
Example 234
Nl,N4-bis(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-
20 yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)terephthalamide
Figure imgf000365_0001
Compound 234, Nl,N4-bis(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethyl)- terephthalamide: Compound 234 was prepared following the procedures outlined in Example 215 using terephthalic acid (13.8 mg, 0.0833 mmol) and N-(2-(2-(2- aminoethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)benzenesulfonamide (Compound 168.2, 121.7mg, 0.167mmol). Purification by preparative HPLC gave the title compound (60.0mg) as a TFA salt. 1H-NMR (400MHz, CD3OD): δ 7.88 (m, 6H), 7.72 (s, 2H), 7.61 (t, 2H), 7.51 (m, 4H), 6.80 (s, 2H), 4.88-4.75 (m, 4H), 4.75 (d, 2H), 4.74 (m, 2H), 3.85-3.42 (m, 25H), 3.12 (s, 6H), 2.99 (t, 4H). MS (m/z): 1135.11 [M+H]+.
Example 235 N,N'-(10-oxo-3,6,14,17-tetraoxa-9,ll-diazanonadecane-l,19-diy])bis(3-(6,8- dichloro-2-methy]-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesu]fonamide)
Figure imgf000365_0002
Compound 235, N,N'-(10-oxo-3,6,14,17-tetraoxa-9,ll-diazanonadecane-l,19- diyl)bis(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)benzenesulfonamide): Compound 235 was prepared following the procedures S outlined in Example 232 using N-(2-(2-(2-ammoethoxy)ethoxy)ethyl)-3-(6,8-dichloro- 2-methyl-l,2,3,4-tetrahydroisoqumolm-4-yl)benzenesulfonamide (Compound 168 2, 56 6mg, 00775mmol) Purification by preparative HPLC gave the title compound (25 Omg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 88 (d, 2H), 7 75 (s, 2H, 7 65 (t, 2H), 7 53 (m, 4H), 6 83 (s, 2H), 4 89-4 68 (m, 2H), 3 88 (m, 2H), 3 62-3 43 (m, 10 21H), 3 30-3 27 (m, 6H), 3 11 (s, 7H), 3 03 (t, 4H) MS (m/z) 1031 07 [M+H]+
Example 236 N,N'-(10,17-dioxo-3,6,21,24-tetraoxa-9,ll,16,18-tetraazahexacosane-l,26- diyl)bis(3-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- 15 yl)benzenesulfonamide)
Figure imgf000366_0001
Compound 236, N,N'-(10,17-dioxo-3,6,21,24-tetraoxa-9,l 1,16,18-
20 tetraazahexacosane-l,26-diyl)bis(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide): Compound 236 was prepared following the procedures outlined in Example 208 using 1 ,4-diisocyanatobutane (5 24mg, 0 0374mmol) and N-(2-(2-(2-ammoethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoqumolm-4-yl)benzenesulfonamide (Compound 168 2, 25 54 7mg, 0 0749mmol) Purification by preparative HPLC gave the title compound (27 5mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 88-7 86 (d, 2H), 7 75 (s, 2H), 7 63 (t, 2H), 7 55-7 51 (m, 4H), 4 48 (m, 2H), 3 38-3 31 (m, IH), 3 61-3 42 (m, 17H), 3 35-3 30 (m, 4H), 3 13 (s, 6H), 3 08-3 02 (m, 7H), 1 45 (m, 2H) MS (m/z) 1145 04 [M+H]+
Example 237 N,N'-(2,2'-(2,2'-(2,2'-(l,4- phenylenebis(azanediyl))bis(oxomethylene)bis(azanediyl)bis(ethane-2,l- diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(3-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide)
Figure imgf000367_0001
Compound 237, N,N'-(2,2'-(2,2'-(2,2'-(l,4- phenylenebis(azanediyl))bis(oxomethylene)bis(azanediyl)bis(ethane-2,l- diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(3-(6,8-dichloro-2- methyI-l,2,3,4-tetrahydroisoquinolin-4-yI)benzeπesulfonamide): Compound 237 was prepared following the procedure outlined in Example 208 using 1,4- dnsocyanatobenzene (8 79mg, 0 0549mmol) and N-(2-(2-(2- ammoethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l ,2,3,4 tetrahydroisoqumohn 4- yl)benzenesulfonamide (Compound 168 2, 80 2mg, O l lOmmol) Puπfication by preparative HPLC gave the title compound (37 6mg) as a TFA salt 1H-NMR (400 MHz, CD3OD) δ 7 88 (d, 2H), 7 73 (s, 2H), 7 61 (t, 2H), 7 52 (d, 2H), 7 48 (d, 2H), 7 18 (s, 5H), 6 78 (s, 2H), 4 71-4 63 (m, 6H), 4 45-4 40 (m, 2H), 3 81-3 77 (m, 2H), 3 58-3 55 (m, 6H), 3 53-3 50 (m, 14H), 3 47-3 44 (m, 6H), 3 35-3 33 (m, 6H), 3 09 (s, 8H), 3 03 (t, 5H) MS (m/z) 1 165 06 [M+H]+ Example 238
N,N'-(10,17-dioxo-3,6,21,24-tetraoxa-9,ll,16,18-tetraazahexacosane-l,26- diyl)bis(4-(6,8-dichIoro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)benzenesulfonamide)
Figure imgf000368_0001
Compound 238, N,N'-(10,17-dioxo-3,6,21,24-tetraoxa-9,ll,16,18- tetraazahexacosane-l,26-diyl)bis(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide): Compound 238 was prepared following the procedure outlined m Example 208 using 1 ,4-diisocyanatobutane (5 64mg, 0 402mmol) and N-(2-(2-(2-ammoethoxy)ethoxy)ethyl)-4-(6)8-dichloro-2- methyl-l,2,3,4-tetrahydroisoquinohn 4-yl)benzenesulfonamide (Compound 175 1, 58 8mg, 0 805mmol) Puπfication by preparative HPLC gave the title compound (13 8 mg) as a TFA salt 1H-NMR (400 MHz, CD3OD) δ 7 86 (d, J-8Hz, 2H), 7 72 (s, 2H), 7 61 (t, 2H), 7 52 (s, 2H), 747 (d, J"7Hz, 2H), 7 18 (s, 5H), 7 78 (s, 2H), 4 77-4 68 (m, 5H), 448-4 40 (m, 2H), 3 35-3 28 (m, 2H), 3 56-3 51 (m, 16H), 3 45 (t, J-5Hz, 5H), 3 35-3 32 (m, 10H), 3 09 (s, 6H), 3 03 (t, J 5Hz, 3H) MS (m/z) 1145 01 [M+H]+
Example 239
N,N'-(2,2'-(2,2'-(2,2'-(l,4- phenylenebis(azanediyl))bis(oxomethylene)bis(azanediyl)bis(ethane-2,l- diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(4-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide)
Figure imgf000369_0001
Compound 239, N,N'-(2,2'-(2,2'-(2,2'-(l,4- phenylenebis(azanediyl))bis(oxomethylene)bis(azanediyl)bis(ethane-2,l- diyl))bis(oxy)bis(ethane-2,l-diyl))bis(oxy)bis(ethane-2,l-diyl))bis(4-(6,8-dichloro-2- methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)beπzenesulfonamide): Compound 239 was prepared following the procedure outlined in Example 208 using 1,4- dnsocyanatobenzene (12 5 mg, 0 078 mmol) and N-(2-(2-(2- ammoethoxy)ethoxy)ethyl)-4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinohn-4- yl)benzenesulfonamide (Compound 175 1, 113 9 mg, 0 156 mmol) Purification by preparative HPLC gave the title compound (48 9 mg) as a TFA salt 1H-NMR (400 MHz, CD3OD) δ 7 87 (d, J-8Hz, 4H), 7 52 (s, 2H), 7 40 (d, J=8Hz, 4H), 7 18 (s, 4H), 7 69 (s, 2H), 4 70-4 62 (m, 3H), 4 48-4 40) (m, 2H), 3 82-3 76 (m, 2H), 3 58-3 43 (m, 21H), 3 35-3 30 (m, 4H), 3 11-3 06 (m, 1 IH) MS (m/z) 1165 12[M+H]+
Example 240 (2S,3S)-Nl,N4-bis(2-(2-(2-(2-(3-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccin amide
Figure imgf000370_0001
Compound 240, (2S,3S)-Nl,N4-bis(2-(2-(2-(2-(3-((S or R)-6,8-dichloro-2-methyl- l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide:
Compound 240 was prepared following the procedures outlined in Example 229 using (2S,3S)-2,3-dihydroxysuccinic acid (9 6mg, 0 057mmol) and (S or R)-N-(2-(2-(2-(2- aminoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoquinolm-4-yl)benzenesulfonamide (intermediate 224 1, 88 6mg,
10 0 114mmol) Purification by preparative HPLC gave the title compound (24 5mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 94 (t, IH), 7 87 (d, 2H), 7 77 (s, 2H), 7 63 (t, 2H), 7 53-7 50 (m, 4H), 6 82 (s, 2H), 4479-4 45 (m, 2H), 444 (s, 2H), 3 88-3 84 (m, 2H), 3 62-3 53 (m, 22H), 3 50-3 48 (m, 5H), 3 45-3 40 (m, 9H), 3 13 (s, 6H), 3 04 (t, 4H) MS (m/z) 1208 02 [M+H]+
I5
Example 241
(2R,3R)-Nl,N4-bis(2-(2-(2-(2-(3-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinoHn-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide
20
Figure imgf000371_0001
Compound 241, (2R,3R)-Nl,N4-bis(2-(2-(2-(2-(3-((R or S)-6,8-dichloro-2-methyl- l,2,3,4-tetrahydroisoquinolin-4- yl)pheny]sulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide:
Compound 241 was prepared following the procedures outlined in Example 229 using (2R,3R)-2,3-dihydroxysuccinic acid (8 7mg, 0 0519mmol) and (S or R)-N-(2-(2-(2-(2- ammoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoqumolm-4-yl)benzenesulfonamide (intermediate 224 1, 80 5mg,
10 0 104mmol) Puπfication by preparative HPLC gave the title compound (25 7) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 87 (d, 3H), 7 76 (s, 2H), 7 63 (t, 2H), 7 54-7 51 (m, 4H), 6 83 (s, 2H), 4 78-4 73 (m, 4H), 449-442 (m, 4H), 3 89-3 85 (m, 2H), 3 62-3 53 (m, 22H), 3 51-48 (m, 5H), 3 46-3 38 (m, 9H), 3 14 (s, 6H), 3 04 (t, 4H) MS (mA) 1208 21 [M+H]+
I5
Example 242
(2S,3S)-Nl,N4-bis(2-(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide
20
Figure imgf000372_0001
Compound 242, (2S,3S)-Nl,N4-bis(2-(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyl- l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysuccinamide:
Compound 242 was prepared following the procedures outlined in Example 229 using (2S,3S)-2,3-dihydroxysuccimc acid (6 3mg, 0 0374mmol) and (S or R)-N-(2-(2-(2-(2- ammoethoxy)ethoxy)ethoxy)ethyl)-4-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoqumolm-4 yl)benzenesulfonamide (intermediate 225 2, 58 Omg, 00749mmol) Puπflcation by preparative HPLC gave the title compound (21 6mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 85 (d, 4H), 7 54 (s, 2H), 7 45 (d, 3H), 6 84 (s, IH), 4 772-4 69 (m, 3H), 4 43 (s, 2H), 3 86-3 81 (m, IH), 3 59-3 53 (m, 16H), 3 49-3 39 (m, 1 IH), 3 12 (s, 5H), 3 08 (t, 4H) MS (m/z) 1208 14 [M+H]+
Example 243
(2R,3R)-Nl,N4-bis(2-(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3- dihydroxysuccinamide
Figure imgf000373_0001
Compound 243, (2R,3R)-Nl,N4-bis(2-(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyl- l,2,3,4-tetrahydroisoquinolin-4- yl)phenyIsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)-2,3-dihydroxysucclnamide:
Compound 243 was prepared following the procedures outlined in Example 229 using (2R,3R)-2,3-dihydroxysuccimc acid (8 4mg, 0 0 0499mmol) and (S or R)-N-(2-(2-(2- (2-ammoethoxy)ethoxy)ethoxy)ethyl)-4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoqumohn-4-yl)benzenesulfonamide (intermediate 225 2, 77 3 mg, 0 0999 mmol) Purification by preparative HPLC gave the title compound (23 4mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 89 (d, 4H), 7 53 (s, 2H), 7 45 (d, 4H), 6 83 (s, 2H), 4 81-4 71 (m, 4H), 4 49-4 41 (m, 4H), 3 89-3 83 (m, 2H), 3 60-3 53 (m, 17H), 3 49-3 38 (m, 12H), 3 13 (s, 5H), 3 08 (t, 4H) MS (m/z) 1208 09 [M+H]+
Example 244
(S or R)-N,N'-(13,20-dioxo-3,6,9,24,27,30-hexaoxa-12,14,19,21- tetraazadotriacontane-l,32-diyl)bis(3-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquiπolin-4-yl)benzenesulfonamide)
Figure imgf000373_0002
Intermediate 244.1, (S or R)-4-(3-bromophenyl)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinoline: Into a 2000-mL round-bottom flask, was placed a solution of 4-(3-bromophenyl)-6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolme (intermediate 5 1 4, 20 g, 5420 mmol, 1 00 equiv) in ethanol (500 mL) This was followed by the addition of D-(+)-dibenzoyl tartaric acid (19 g, 53 07 mmol, 0 98 equiv), water (160 mL) and ethanol (1440 mL) at 450C The resulting solution was stirred for 30 min at 450C in an oil bath After cooling to room temperature over 24 hours, the solids were collected by filtration The filter cake was dissolved m potassium carbonate (saturated )Q and was extracted with 2x500 mL of ethyl acetate The combined organic layers were washed with 2x500 mL of bπne, dried over anhydrous sodium sulfate and concentrated under vacuum This gave (S or R)-4-(3-bromophenyl)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoqumolme as a colorless oil
Figure imgf000374_0001
Intermediate 224.1 (alternate synthesis), (S or R)-N-(2-(2-(2-(2- aminoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide (S or R)-4-(3-bromophenyl)-6,8-Q dichloro-2-methyl-l,2,3,4-tetrahydroisoquinohne (intermediate 244 1) was converted to (S or R)-N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl- l,2,3,4-tetrahydroisoquinohn-4-yl)benzenesulfonamide (intermediate 224 1) following the procedures outlined for the racemic substrates in Example 1 and the reduction described in Example 170 5
Figure imgf000375_0001
Compound 244, (S or R)-N,N'-(13,20-dioxo-3,6,9,24,27,30-hexaoxa-12,14,19,21- tetraazadotriacontane-l,32-diyl)bis(3-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide): Compound 244 was prepared following the procedures outlined in Example 208 using 1 ,4-diisocyanatobutane (6.5mg, 0.0471mmol) and (S or R)-N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-3- (6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4-yl)benzenesulfonamide (Intermediate 224.1, 72.9mg, 0.0941mmol). Purification by preparative HPLC gave the
10 title compound (34.9mg) as a TFA salt. 1H-NMR (400MHz, CD3OD): δ 7.89 (d, 2H), 7.75 (s, 2H), 7.63 (t, 2H), 7.55-7.51 (m, 4H), 6.83 (s, 2H), 4.48 (d, 2H), 3.90-3.85 (m, 2H), 3.59-3.55 (m, 17H), 3.51-3.43 (m, 14H), 3.31-3.23 (m, 6H), 3.14 (s, 7H), 3.04 (m, 9H), 1.43 (m, 4H). MS (m/z): 1232.99 [M+H]+.
I5 Example 245
(S or R)-N,N'-(l,l'-(l,4-phenylenebis(azanediyl))bis(l-oxo-5,8,ll-trioxa-2- azatridecane-13,l-diyl))bis(3-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yI)benzenesulfonamide)
Figure imgf000375_0002
Compound 245, (S or R)-N,N'-(l,l'-(M-phenylenebis(azanediyl))bis(l-oxo-5,8,ll- trioxa-2-azatridecane-13,l-diyl))bis(3-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide): Compound 245 was prepared following the procedures outlined in Example 208 using (S or R)-N-(2-(2-(2-(2- aminoethoxy)ethoxy)emoxy)ethyl)-3-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide (Intermediate 224.1, 79.1mg, 0.102mmol) and 1,4-diisocyanatobenzene (8.2mg, 0.051 lmmol). Purification by preparative HPLC gave the title compound (43.2mg) as a TFA salt. 1H-NMR (400MHz, CD3OD): δ 7.87 (d, 2H), 7.72 (s, 2H), 7.61 (t, 2H), 7.51-7.46 (m, 4H), 7.17 (s, 4H), 6.78 (s, 2H), 4.44-4.39 (m, 2H), 3.82-3.77 (m, 2H), 3.61 (s, 1 IH), 3.57-3.53 (m, 13H), 3.49-3.48 (m, 6H), 3.44 (t, 5H), 3.35-3.29 (m, 6H), 3.09 (s, 7H), 3.03 (t, 4H). MS (Wz): 1253.01 [M+H]+.
Compound 246
Nl,N4-bis(2-(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsuIfonamido)ethoxy)ethoxy)ethoxy)ethyI)- terephthalamide
Figure imgf000376_0001
Compound 246, Nl,N4-bis(2-(2-(2-(2-(4-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)- terephthalamide: Compound 246 was prepared following the procedures outlined in Example 215 using (S or R)-N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-4-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoqumolm-4-yl)benzenesulfonamide (Intermediate 224 1, 65 lmg, 0 0841mmol) and terephthalic acid (698mg, 0 042mmol) Purification by preparative HPLC gave the title compound (193mg) as a TFA salt 1H- NMR (400MHz, CD3OD) δ 7 89-7 85 (m, 6H), 7 52 (s, 2H), 743 (d, 4H), 6 81 (s, 2H), 4 73-4 66 (m, 3H), 447-442 (m, IH), 3 84-3 79 (m, 2H), 3 64-3 59 (m, 14H), 3 57-3 54 (m, 1 IH), 3 46-3 39 (m, 8H), 3 12 (s, 6H), 3 03 (t, 4H) MS (m/z) 1233 04 [M+H]+
Example 247
Nl-(2-(2-(2-(2-(3-(6,8-dichloro-2-methy]-l,2,3,4-tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)succinamide
Figure imgf000377_0001
Compound 247, Nl-(2-(2-(2-(2-(3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4- yl)phenylsulfonamido)ethoxy)ethoxy)ethoxy)ethyl)succinamide: Compound 247 was prepared following the procedure outlined in Example 215 using 4-ammo-4- oxobutanoic acid (7 6 mg, 00646 mmol) and N-(2-(2-(2-(2- ammoethoxy)ethoxy)ethoxy)ethyl)-3-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoqumolin-4-yl)benzenesulfbnamide (Compound 28, 50 mg, 0 0646 mmol) Purification by preparative HPLC gave the title compound (27 8 mg) as a TFA salt 1H- NMR (400MHz, CD3OD) δ 7 88 (d, IH), 7 75 (s, IH), 7 64 (t, IH), 7 55 (s, IH), 7 51 (d, IH), 6 84 (s, IH), 4 78-4 71 (m, 2H), 4 55-448 (m, IH), 3 81-3 75 (m, IH), 3 63- 3 55 (m, 10H), 3 51-4 45 (m, 5H), 3 44-3 41 (m, 3H), 3 38-3 31 (m, 3H), 3 13 (s, 3H), 3 07-3 02 (t, 2H), 2 48-2 43 (m, 4H) MS (m/z) 645 32 [M+H]+
Example 248
N,N'-(13,20-dioxo-3,6,9,24,27,30-hexaoxa-12,14,19,21-tetraazadotriacontane-l,32- diyl)bis(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)benzenesulfonamide)
10
Compound 248, N,N'-(13,20-dioxo-3,6,9,24,27,30-hexaoxa-12,14,19,21- tetraazadotriacontane-l,32-diyl)bis(4-(6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yI)benzenesulfonamide): Compound 248 was prepared following the procedure outlined in Example 208 using 1,4-diisocyanatobutane (7 64
15 mg, 0 545 mmol) and N-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-4-(6,8- dichloro-2-methyl-l,2,3,4-tetrahydroisoquinohn-4-yl)benzenesulfonamide (Compound 82, 844 mg, 0 109mmol) Purification by preparative HPLC gave the title compound (43 6mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 89 (d, 4H), 7 54 (s, 2H), 7 45 (d, 4H), 6 84 (s, 2H), 4 79 4 71 (m, 4H), 3 89-3 85 (dd, 2H), 3 59-3 56 (m, 17H),
20 3 49-3 43 (m, 14H), 3 28-3 23 (m, 5H), 3 14 (s, 7H), 3 09 3 04 (m, 9H), 1 42 (s, 4H) MS (m/z) 1233 03 [M+H]+
Example 249 N^V'-(l,l'-(l,4-phenyIenebis(azanediyl))bis(l-oxo-5,8,ll-trioxa-2-azatridecane-
13,l-diyl))bis(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)benzenesulfonamide)
Figure imgf000379_0001
Compound 249, N,N'-(l,l'-(l,4-phenylenebis(azanediyl))bis(l-oxo-5,8,ll-trioxa-2- azatridecane-13,l-diyl))bis(4-(6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin- 4-yl)benzenesulfonamide): Compound 249 was prepared following the procedure outlined in Example 208 using 1 ,4-diisocyanatobenzene (7 95mg, 0 0495mmol) and N- (2-(2 (2 (2-ammoethoxy)ethoxy)ethoxy)ethyl)-4-(6,8-dichloro-2-methyl-l ,2,3,4- tetrahydroisoqmnolin-4-yl)benzenesulfonamide (Compound 82, 76 7mg, 0 099mmol) Puπfication by preparative HPLC gave the title compound (39 6 mg) as a TFA salt 1H- NMR (400MHz, CD3OD) δ 7 87 (d, 4H), 7 51 (s, 2H), 7 40 (d, 4H), 7 16 (s, 4H), 6 79 (s, 2H), 4 88-4 83 (m, 4H), 4 65-4 50 (m, 2H), 3 81-3 77 (m, 2H), 3 61-3 59 (m, 9H), 3 58-3 54 (m, HH), 3 53-3 48 (m, 5H), 3 47-3 42 (m, 5H), 3 35-3 30 (m, 4H), 3 11 (s, 6H), 3 07 (t, 4H) MS (m/z) 1253 04 [M+H]+
Example 250 (S or R)-N,N'-(13-oxo-3,6,9,17,20,23-hexaoxa-12,14-diazapentacosane-l,25- diyl)bis(4-((S or R)-6,8-dichloro-2-methyl-l,2,3,4-tetrahydroisoquinolin-4- yl)benzenesulfonamide)
Figure imgf000380_0001
Compound 250, (S-or R)-N,N'-(13-oxo-3,6,9,17,20,23-hexaoxa-12,14- diazapentacosane-l,25-diyl)bis(4-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesuIfonamide): Compound 250 was prepared following the procedures outlined in Example 232 using (S or R)-N-(2-(2-(2-(2- aminoethoxy)ethoxy)ethoxy)ethyl)-4-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoquiπoliπ-4-yl)benzenesulfonamide (Intermediate 225 2, 75 mg, 0 0968mmol) Purification by preparative HPLC gave the title compound (26 0 mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 88 (d, 4H), 7 54 (s, 2H), 7 45 (d, 4H), 6 84 (s, 2H), 4 79-4 72 (m, 5H), 448-4 42 (m, 2H), 3 87-3 83 (m, 2H), 3 58-3 54 (m, 17H), 3 49-3 43 (m, 15H), 3 24-3 22 (m, 6H), 3 12 (s 6H), 3 08 (t, 4H) MS (m/z) 1118 96 [M+H]+
Example 251
(S or R)-N,N'-(13,20-dioxo-3,6,9,24,27,30-hexaoxa-12,14,19,21- tetraazadotriacontane-l,32-diyl)bis(4-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide)
Figure imgf000381_0001
Compound 251, (S or R)-N,N'-(13,20-dioxo-3,6,9,24,27,30-hexaoxa-12,14,19,21- tetraazadotriacontane-l,32-diyl)bis(4-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide): Compound 251 was prepared following the procedures outlined in Example 208 using (S or R)-N-(2-(2-(2-(2- aminoethoxy)ethoxy)ethoxy)ethyl)-4-(6,8-dichloro-2-methyl-l, 2,3,4- tetrahydroisoquinolm-4-yl)benzenesulfonamide (intermediate 225 2, 88 lmg, 0 114mmol) and 1,4-diisocyanatobutane (7 9mg, 0 0569mmol) Purification by preparative HPLC gave the title compound (56 lmg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 85 (d, 4H), 7 54 (s, 2H), 745 (d, 4H), 6 84 (s, 2H), 4 77-4 74 (m, 4H), 4 50-4 46 (m, 2H), 3 89-3 84 (m, 2H), 3 61-3 56 (m, 17H), 3 50-3 43 (m, 14H), 3 26-3 23 (m, 6H), 3 14 (s, 7H), 3 09-3 04 (m, 10H), 1 48 (s, 4H) MS (m/z) 1233 01 [M+H]+
Example 252
(S or R)-N,N'-(l,l'-(l,4-phenylenebis(azanediyl))bis(l-oxo-5,8,ll-trioxa-2- azatridecane-13,l-diyl))bis(4-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yl)benzenesulfonamide)
Figure imgf000382_0001
Compound 252, (S or R)-N,N'-(l,l'-(l,4-phenylenebis(azanediyl))bis(l-oxo-5,8,ll- trioxa-2-azatridecane-13,l-diyl))bis(4-((S or R)-6,8-dichloro-2-methyl-l,2,3,4- tetrahydroisoquinolin-4-yI)benzenesulfonamide): Compound 252 was prepared following the procedures outlined in Example 208 using (S)-N-(2-(2-(2-(2- aminoethoxy)ethoxy)ethoxy)ethyl)-4-(6,8-dichloro 2-methyl-l, 2,3,4- tetrahydroisoqumohn-4-yl)benzenesulfonamide (intermediate 225 2, 45 2mg, 00584mmol) and 1,4-diisocyanatobenzene (4 7 mg, 0 0292 mmol) Purification by preparative HPLC gave the title compound (20 7 mg) as a TFA salt 1H-NMR (400MHz, CD3OD) δ 7 87 (d, 4H), 7 51 (s, 2H), 7 39 (d, 4H), 7 16 (s, 4H), 6 79 (s, 2H), 4 72-4 61 (m, 4H), 4 46-3 99 (m, IH), 3 81 3 73 (m, IH), 3 62-3 42 (m, 33H), 3 35-3 33 (m, 5H), 3 09-3 06 (m, 13H) MS (Wi) 1252 95 [M+H]+
Topological Polar Surface Area Data
Topological Polar Surface Area (tPSA) values for representative compounds in the disclosure are shown in Table 7, below The tPSA values were calculated using the method of Ertl et al , Journal of Medicinal Chemistry, 43 3714 3717 (2000)
Table 7 tPSA Values of Compounds
Topological polar
Example # surface area (A2)
Figure imgf000383_0001
Figure imgf000384_0001
Figure imgf000385_0001
Figure imgf000386_0001
Figure imgf000387_0001
Figure imgf000388_0001
1 Pharmacological Test Example 1 Cell-based assay of NHE 3 activity Rat NHE-3 -mediated Na+- dependent H+ antiport was measured using a modification of the pH sensitive dye method originally reported by Tsien (Proc Natl Acad Sa U S A (1984) 81(23) 7436-7440) Opossum kidney (OK) cells were obtained from the ATCC and propagated per their instructions The rat NHE-3 gene was introduced into OK cells via electroporation, seeded into 96 well plates and grown overnight Medium was aspirated from the wells, cells were washed twice with NaCl-HEPES buffer (100 mM NaCl, 50 mM HEPES, 10 mM glucose, 5mM KCl, 2 mM CaCl2, 1 mM MgCl2, pH 7 4), then incubated for 30 mm at room temperature with NH4Cl-HEPES buffer (20 mM NH4Cl, 80 mM NaCl, 50 mM HEPES, 5 mM KCl, 2mM CaCl2, 1 mM MgCl2, pH 7 4) containing 5 uM BCECF-AM (Invitrogen) Cells were washed twice with Ammonium free, Na+-free HEPES (100 mM choline, 50 mM HEPES, 10 mM glucose, 5 mM KCl, 2 mM CaCk, 1 nαM MgCk, pH 7 4) and incubated in the same buffer for 10 minutes at room temperature to lower intracellular pH NHE-3-mediated recovery of neutral intracellular pH was initiated by addition of Na-HEPES buffer containing 5 uM ethyl isopropyl amiloπde (EIPA, a selective antagonist of NHE-I activity that does not inhibit NHE-3) and 0-30 uM test compound, and monitoring the pH sensitive changes in BCECF fluorescence Q^x 505nm, λem 538nm) normalized to the pH insensitive BCECF fluorescence (λex 439nm, λem 538nm) Initial rates were were plotted as the average 3-6 replicates, and pIC50 values were estimated using GraphPad Pπsm The inhibitory data of many of the example compounds illustrated above are shown in Table 8, below
Figure imgf000389_0001
Figure imgf000390_0001
Figure imgf000391_0001
1 pIC50 is the negative log the IC50 value (an IC50 value of 1 micromolar corresponds to a pIC50 value of6 0)
2 Pharmacological Test Example 2 Parallel Artificial Membrane Permeability Assay (PAMPA) The model consists of a hydrophobic filter mateπal coated with a mixture of lecithin/ phospholipids creating an artificial lipid membrane BD Gentest PAMPA 96-well plates (cat #353015) are warmed for 1 hr at room temperature 1 mL of 20 uM control compounds (pooled mix of 10 mM atenolol, ranitidine, labetalol, and propranolol) in transport buffer (10 mM HEPES in HBSS pH 7 4) are prepared along with 1 mL of 20 uM test compounds in transport buffer The PAMPA plates are separated, and 0 3 mL of compound are added in duplicate to apical side (bottom/donor plate- "AP"), and 2 mL buffer are placed in the basolateral chamber (top/receiver plate="BL") The BL plate is placed on theAP plate and incubated for 3 hrs in 37°C incubator At that time, samples are removed from both plates, and analyzed for compound concentration using LC/MS A "Pe" (effective permeability) value is calculated using the following formula
Pe- (-ln[l-CA(t)/Ceq])/[A*(l/VD+l/VA)*t where
CA = concentration in acceptor well, CD ~~ concentration in donor well VD = donor well volume (mL), VA = acceptor well volume (mL)
A = filter area = 0 3 cm2, t = transport time (seconds) Ceq = equilibrium concentration - [CD(t)*VD+CA(t)*VA]/(VD+VA) Pe is reported in units of cm/sec x 106
Results from PAMPA testing are shown in Table 9
Figure imgf000392_0001
Figure imgf000393_0001
Increasing values of tPSA are typically associated with lower permeability. Figure 1 illustrates the Relationship between tPSA and Permeability (Papp, as measured in the PAMPA assay) of Example compounds Compounds with higher tPSA values trend toward lower permeability
3 Pharmacological Test Example 3 Pharmacodynamic Model Effect of test compounds on fluid content of intestinal compartments Normal female Sprague Dawley rats, 7 weeks old, were acclimated for at least 2 days The animals were fed ad lib through the experiment Groups of 5 rats were orally gavaged with 1 5 mL of water containing a negative control compound or test compounds, adjusted to a concentration that results m a dose of lO mg/kg Six hours after dosing, rats were euthanized with isofluorane The cecum and colon were ligated and then removed After a bnef πnse in saline and pat-drying, the segments were weighed The segments were then opened, and the contents collected and weighed The collected contents were then dπed, and weighed again The %water content was reported as 100 x ((Ww - Wd) / Ww) where Ww is the weight of the wet contents, and Wd is the weight of the contents after drying The differences between groups are evaluated by one way ANOVA with Bonferroni post tests Examples are shown in Figures 2 A and 2B (wherein rats were dosed orally with 10 mg/kg of compound (Example or Control), and then after 6 hours, cecum and colon contents were removed, weighed and dπed, and the % water in the contents was determined *, P < 0 05 and ***, P < 0 01 compared to control in ANOVA analysis)
4 Pharmacological Test Example 4
Determination of compound Cmax and AUC Sprague-Dawley rats were orally gavaged with test article (2 5 mg/kg) and serum was collected at 0 5, 1, 2 and 4 h Serum samples were treated with acetomtπle, precipitated proteins removed by centπfugation and supernatants analyzed by LC/MS/MS and compared against a standard curve to determine compound concentration Table 10 illustrates data from the pharmacokinetic profiling of selected example compounds All compounds were orally dosed at the dosage shown, and pharmacokinetic parameters determined as descπbed in the text
Figure imgf000395_0001
Figure imgf000396_0001
5 Pharmacological Test Example 5
Evaluation of NHE-3 -inhibitory Compounds in Disease Models with Na/H,O Retention CRF/ESRD Model Male Sprague Dawley rats with subtotal (5/6th) nephrectomy, 7 weeks old and weighing 175-200 g at surgery time, are purchased from Charles River Laboratories The animals are subjected to acclimation for 7 days, and randomly grouped (using random number table) before proceeding to experiments During acclimation, all animals are fed with base diet HD8728CM The rats are housed in holding cages (2/cage) duπng the acclimation peπod and the time between sample collections The rats are transferred to metabolic cages on the days of sample collections Food and water is provided ad libitum
Chrome renal failure is induced in the rats by subtotal (5/6th) nephrectomy (Nx) followed by intravenous (IV) injection of adπamycin (ADR) at 2 weeks post-nephrectomy, at a dose of 3 5mg/kg body weight Animals are then randomized into control and treatment groups with 10 rats per group Rats in untreated group are fed with base diet and rats in the treatment groups are fed the same chow supplemented with NHE-3 inhibitor/fluid holding polymer at various doses All the groups are maintained for 28 days Serum samples are collected at day (-1) (1 days before ADR injection), days 14 and 28 post ADR treatment Twenty four hour uπne and fecal samples are collected at day (-1), days 14 and 28 post ADR treatment and stored at -2O0C for later analysis Body weight, food and water consumption are measured at the same time points as uπne collections Serum and urme chemistry (Na, K, Ca, Cl) are determined using an ACE Clinical Chemistry System (ALFA WASSER MANN Diagnostic Technologies, LLC) Fecal electrolyte (Na, K, Ca, Cl) excretions are determined by IC Fluid balance are also determined via amount of fluid intake (in drinking water) subtracted by combined fecal water amount and uπne volume Tissues (heart, kidney and small intestine) are harvested at the end of expeπments for later histopathological analysis The third space (pleural fluids and ascites) body fluid accumulation are scored semi-quantitatively as follows grade 0, no fluid accumulation, grade 1 , trace amount of fluids, grade 2, obvious amount of fluids, grade 3, both cavities full of fluids, grade 4, fluids overflowed once the cavities are opened Each score of body fluid accumulation is confirmed and agreed on by 2 investigators Animals treated with NHE 3 inhibitor/fluid holding polymer show decreased serum aldosterone, decreased 24 hr uπne volume and decreased uπne K excretion, and increased unne Na excretion compared to no treatment group Treated animals also have increased fecal Na and fluid excretion, compared to control group Compared to untreated rats which show positive fluid balance of 4 g per day, animals treated with NHE-3 mhibitor/fluid holding polymer demonstrate a fluid loss of 5 g per day Treatment of NHE-3 inhibitor/fluid holding polymer in CRF rats is associated with less edema m heart, kidney and small intestine tissues, less hypertrophy in heart, less third space fluid accumulation, and lower body weight at the end of experiment compared to untreated group
6 Pharmacological Test Example 6
Evaluation of NHE-3 -inhibitory Compounds in Disease Models with Na/H2O Retention Congestive Heart Failure Model CHFs are introduced to male Spraque Dawley rats, 7 -8 weeks old fed ad lib regular diet and ad lib 10% ethanol m drinking water, and gavaged with a daily dose of 6 3 mg cobalt acetate for 7 days Then CHF rats are gavaged with a daily dose of 4 mg of furosemide for 5 days, inducing resistance to furosemide diuretic effects The rats are then randomly divided into 2 groups, control and treatment, and the treatment group admistered NHE-3 inhibitor/fluid holding polymer for 7 days Day 0 and day 7 post treatment serum aldosterone levels, uπne volume, uπne Na and K excretions are measured Fluid balance is also determined via amount of fluid intake (in drinking water) subtracted by combined fecal fluid amount and uπne volume
Animals treated with NHE 3 inhibitor/fluid holding polymer have decreased serum aldosterone levels, decreased 24hr uπne volume and uπne K excretion, and increased uπne Na excretion compared to control group Animals treated with NHE-3 inhibitor/fluid holding polymer have, for example, increased fecal Na and fluid excretion Compared to untreated rats, which show a positive fluid balance of, for example, 4 g per day, treated animals demonstrate a fluid loss of 5 g per day
7 Pharmacological Test Example 7
Evaluation of NHE-3-ιnhιbιtory Compounds in Disease Models with
Na/H2O Retention Hypertension Model Male Dahl salt-sensitive rats are obtained from Harlan Teklad After acclimation, animals are randomly grouped and fed diet containing 8% NaCl ± NHE-3 mhibitor/fluid holding polymer for 7 days Day 0 and day 7 post treatment systolic BP, serum aldosterone levels, urine volume, unne Na and K excretions are measured Fluid balance is also determined via amount of fluid intake (m dπnking water) subtracted by combined fecal fluid amount and urine volume
Animals treated with NHE-3 inhibitor/fluid holding polymer would show decreased systolic BP, serum aldosterone levels, 24 hr urine volume and urine K 5 excretion, and increased urine Na excretion compared to no treatment group Animals treated with NHE-3 inhibitor/fluid holding polymer would also show increased fecal fluid excretion Compared to untreated rats which would show positive fluid balance of 4 g per day, animals treated with NHE-3 inhibitor/fluid holding polymer demonstrate a fluid loss of 2 g per day 10
8 Pharmacological Test Example 8
Na Transport Inhibition Study on Colonic Tissues Immediately following euthanasia and exsanguinations of the rats, the entire distal colon is removed, cleansed in ice cold isotonic saline, and partially stπpped of the serosal musculaπs
15 using blunt dissection Flat sheets of tissue are mounted in modified Ussing chambers with an exposed tissue area of 0 64 cm2 Transepithehal fluxes of 22Na+(Perkin Elmer Life Sciences, Boston, MA) are measured across colonic tissues bathed on both sides by 10 ml of buffered saline (pH 74) at 37°C and circulated by bubbling with 95% O2 5% CO2 The standard saline contains the following solutes (in mmol/1) 139 4 Na+, 5 4 K,
20 1 2 Mg2+, 123 2 Cl , 21 0 HCO3 , 1 2 Ca2+, 0 6 H2PO4 , 2 4 HPO2 , and 10 glucose The magnitude and direction of the net flux (Jnet Na) is calculated as the difference between the two unidirectional fluxes (mucosal to serosal, Jms Na and serosal to mucosal, Jsm Na) measured at 15-min intervals for a control period of 45 mm (Per I), under short circuit conditions In some series, Per I is followed by a second 45 min flux peπod (Per
25 II) to determine the acute effects of NHE inhibitors
9 Pharmacological Test Example 9
Pharmacodynamic Model effect of test compounds and FAP on consistency and form of rat stools Normal rats are given a NHE-3 inhibiting
30 compound and optionally a fluid-absorbing or -holding polymer mixed in their diet at escalated doses Distilled water is available at libitum Clinical data monitored are body weight, food intake, water intake, fecal and urinary output Urinary Na, K and creatinine are measured by a Clinical Analyzer (VetAce, Alfa Wassermann Diagnostic Technologies, LLC, West Caldwell, NJ) The consistency of the stools expelled within 24 h after the administration of each drug or vehicle is reported as follows when the feces are unformed, i e , muddy or watery, this is judged to be diarrhea and the percentage diarrhea is reported as the ratio of the number of animals producing unformed stools to the number tested All of the feces is collected just after each evacuation and put into a covered vessel prepared for each animal in order to prevent the feces from drying To investigate the duration of activity of each drug, the feces collected over each 8-h period is dπed for more than 8 h at 700C in a ventilated oven after the wet weight is measured The fecal fluid content is calculated from the difference between the fecal wet weight and the dry weight Fecal Na and K is analyzed by ion Chromatography (Dionex) after acid digestion of the feces specimen
10 Pharmacological Test Example 10
Effect of test compounds and FAP on CKD rats Male Sprague-Dawley rats (275-300 g, Harlan, Indianapolis, IN) are used and have free access to water and Puπna rat chow 5001 at all times A 5/6 nephrectomy is performed to produce a surgical resection CRF model and the treatment study is performed 6 wk after this procedure In one control group, CRF rats are given access to Puπna rat chow, in treated groups, CRF rats are given access to Puπna rat chow mixed with the article, i e a NHE-3 inhibiting compound and optionally a fluid-absorbing or -holding polymer The treatment peπod is 30 days Systolic blood pressure is monitored m all animals with the use of a tail sphygmomanometer (Harvard Apparatus, South Natick, MA) All rats are euthanatized by an intraperitoneal injection of pentobarbital (150 mg/kg body wt), and blood is collected by cardiac puncture for serum Na+(Roche Hitachi Modular P800 chemistry analyzer, Roche Diagnostics, Indianapolis, IN) and creatinine determination (kit 555A, Sigma Chemical, St Louis, MO) Sodium and creatinine is also determined in a unne specimen collected over 24 h immediately before euthanasia
11 Pharmacological Test Example 11 Effect of test compounds on intestinal fluid accumulation in suckling mice Institute of Cancer Research/Harlan Sprague-Dawley (ICR-HSD) suckling mice, 2 to 4 days old (2 l±l 0 g), are dosed orally with 0 1 mL of test solution (vehicle (1 mmol/L HEPES) or NHE inhibitor dissolved in vehicle) After dosing, the mice are kept at room temperature for 3 hours, then killed, the intestinal and body weights measured, and a ratio of the intestinal weight to remaining body weight is calculated A ratio of 0 0875 represents one mouse unit of activity, indicating significant fluid accumulation in the intestine
12 Pharmacological Test Example 12
Determination of Water absorbing Capacity This test is designed to measure the ability of a polymer to absorb 0 9% saline solution against a pressure of 50g/cm2 or 5 kPa The superabsorbent is put into a plastic cylinder that has a screen fabπc as bottom A weight giving the desired pressure is put on top The cylinder arrangement is then placed on a liquid source The superabsorbent soaks for one hour, and the absorption capacity is determined in g/g
This test principle is described in the European Disposables And Nonwovens Association (EDANA) standard EDANA ERT 442 - Gravimetric Determination of Absorption under Pressure or Absorbency Under Load (AUL), or in the AUL test found in column 12 in US patent no 5,601,542, the entire contents of which are incorporated herein by reference for all relevant and consistent purposes Any of these two methods can be used, or the simplified method descπbed below
Equipment • A plastic cylinder having a screen fabnc made of steel or nylon glued to the bottom The fabπc can have mesh openings of 36 μm (designated "400 mesh"), or in any case smaller than the smallest tested particles The cylinder can have an internal diameter of 25 4 mm, and a height of 40 mm A larger cylinder can also be used, such as the apparatus in the EDANA standard ERT 442 - Grαi imetric Determination of Absorption under Pressure • A plastic piston or spacer disc with a diameter slightly smaller than the cylinder's inner diameter For a cup with a 25 4 mm inner diameter the disc can be 25 2 mm wide, 8 mm high, and weigh about 4 4 g
• A weight that exerts a 50 g/cm2 pressure on the superabsorbent (m combination with the piston) For a 25 4 mm inner diameter cylinder (= 5 067 cm2) and a 4 4 g piston, the weight should have a mass of 249 g
• Glass or ceramic filter plate (porosity = 0) The plate is at least 5 mm high, and it has a larger diameter than the cylinder
• Filter paper with a larger diameter than the cylinder Pore size <25 μm • Petri dish or tray
• 0 9% NaCl solution
Procedure
• Put the glass filter plate in a Petri dish, and place a filter paper on top • Fill the Petπ dish with 0 9% NaCl solution - up to the edge of the filter plate
• Weigh a superabsorbent sample that corresponds to a 0 032 g/cm2 coverage on the cylinder's screen fabπc (=0 16 g for a cylinder with a 25 4 mm inner diameter) Record the exact weight of the sample (A) Carefully distπbute the sample on the screen fabπc • Place the plastic piston on top of the distπbuted sample, and weigh the cylinder assembly (B) Then mount the weight onto the piston
• Place the assembly on the filter paper, and let the superabsorbent soak for 60 minutes
• Remove the weight, and weigh the assembly with the swollen superabsorbent (C)
• Calculate the AUL in g/g according to this formula C - B
13 Pharmacological Test Example 13
Pharmacodynamic model effect of test compounds on fecal water content Normal female Sprague Dawley rats (Charles-River laboratoπes international,
Holhster, CA), 7- 8 weeks old with body weight 175 - 200g were acclimated for at least 3 days before proceeding to experiments The animals were provided food (Harlan Teklad 2018c) and water ad lib through the expeπment Animals were randomly grouped with 6 rats per group
The expeπments were initiated by orally dosmg test compounds at 3 mg/kg in volume of 10 ml/kg Rats from control group were gavaged with the same volume of vehicle (water) After dosing, rats were placed in metabolic cages for 16 hrs (overnight) Food and water consumption were monitored After sixteen hours, feces and uπne were collected The percent of fecal water was measured by weighing fecal samples before and after drying
Representative data of % fecal water content are shown in Table 11 (data are expressed as means, with 6 animals per data point) The differences between control and treated groups were evaluated by one way ANOVA with Dunnett post tests Results are significant if p < 0 05
Figure imgf000403_0001
Figure imgf000404_0001
14 Pharmacological Test Example 14
Pharmacodynamic model effect of test compounds on urinary sodium levels It is anticipated that the reduction of absorption of sodium from the intestine will be reflected in reduced levels of sodium in the uπne To test this, the protocols in Example 13 were repeated, but uπne was collected in addition to feces Uπne sodium levels were analyzed by ion chromatography (IC), and the amound of sodium excreted in the uπne was corrected for vaπations in sodium intake by measuπng food consumption In addition, test compounds were administered at several dose levels to demonstrate a dose-response relationship As shown in Figures 3A and 3B for Examples 201, 244, and 260, where as rats excrete about half the sodium they consume in uπne, in rats treated with increasing doses of NHE-3 inhibitor, the amount of sodium excreted in the uπne diminishes significantly and dose dependently
15 Pharmacological Test Example 15
Pharmacodynamic model dose dependent effect of test compound on fecal water content Rats were monitored for fecal water content as m Example 13, and the test compound was administered at several dose levels to demonstrate a dose- response relationship As shown in Figure 4, in rats treated with increasing doses of the NHE-3 inhibitor tested (i e , Example 87), the fecal water content increased significantly and dose dependently
16 Pharmacological Test Example 16
Pharmacodynamic model Addition of a fluid absorbing polymer to chow Rats were monitored for fecal water content as in Example 13, with the addition of a second group that were fed chow with the addition of 1% Psyllium to their diet In addition to fecal water and uπnary sodium, fecal form was monitored on a scale of 1-5, where 1 is a normal pellet, 3 indicates soft and unformed pellets, and 5 indicates watery feces As shown in Figures 5A, 5B and 5C, supplementing the diet with Psyllium resulted in a slight reduction of fecal stool form, but without impacting the ability of the test compound (ι e , Example 224) to increase fecal water content or decrease urinary sodium
17 Pharmacological Test Example 17
Pharmacodynamic model effect of test compounds on acute stress- induced visceral hypersensitivity in female wistar rats Female Wistar rats weighing 220 - 250 g were prepared for electromyography The animals were anaesthetized, and three pairs of mchrome wire electrodes were implanted bilaterally in the stnated muscles at 3 cm laterally from the midline The free ends of electrodes were exteπoπsed on the back of the neck and protected by a glass tube attached to the skin Electromyographic recordings (EMG) were begun 5 days after surgery The electπcal activity of the abdominal stnated muscles were recorded with an electromyograph machine (Mini VIII, Alvar, Pans, France) using a short time constant (0 03 sec ) to remove low-frequency signals (<3 Hz)
Partial restraint stress (PRS), a relatively mild stress, was performed as follows Bnefly, animals were lightly anaesthetized with ethyl-ether, and their freeholders, upper forehmbs and thoracic trunk were wrapped in a confining harness of paper tape to restnct, but not prevent their body movements and placed in their home cage for 2 hours Control sham-stress animals were anaesthetized but not wrapped PRS was performed between 10 00 and 12 00 AM
Colorectal distension (CRD) was accomplished as follows rats were placed in a plastic tunnel, where they were not allowed to move or escape daily dunng 3 consecutive days (3 h /day) before any CRD The balloon used for distension was 4 cm in long and made from a latex condom inserted in the rectum at 1 cm of the anus and fixed at the tail The balloon, connected to a barostat was inflated progressively by steps of 15 mmHg, from 0, 15, 45 and 60 mmHg, each step of inflation lasting 5 mm CRD was performed at T + 2hl5 as a measure of PRS induced visceral hyperalgesia ± test compound or vehicle To determine the antinociceptive effect of test compounds on stress-induced visceral hypersensitivity, test compounds were administered 1 h before CRD in 6 groups of 8 female rats For each parameter studied (the number of abdominal contractions for each 5-min penod during rectal distension) data is expressed as mean ± SEM Comparisons between the different treatments were performed using an analysis of variance (ANOVA) followed by a Dunnett post test The cπtenon for statistical significance is p<0 05 Figure 6 shows the results of this test using the compound illustrated in
Example 224 dosed orally at 10 mg/kg, and shows that at 45 and 60 mm Hg, inhibition of NHE-3 in rats surprisingly reduces visceral hypersensitivity to distension (p < 0 05)
18 Pharmacological Test Example 18 Pharmacodynamic model effect of test compounds on fecal sodium levels It is anticipated that the reduction of absorption of sodium from the intestine will be reflected m increase levels of sodium in the feces To test this, the protocols in Example 13 were repeated After drying of feces to determine water content, IM HCl was added to dπed ground feces to a concentration of 50 mg/mL and extracted at room temperature on rotator for 5 days Sodium content was analyzed by ion chromatography (IC) As shown in Figures 7A and 7B for Example 224, in rats treated with an NHE-3 inhibitor, the amount of sodium excreted in the feces significantly (p < 0 05 by t-test)
19 Pharmacological Test Example 19
Determination of compound remaining in feces Sprague-Dawley rats were orally gavaged with test article A low dose of compound (0 1 mg/kg) was selected so that feces would remain solid and practical to collect For both Examples 202 and 203, three rats were dosed, and following dosage of compounds, the rats were placed in metabolic cages for 72 hours After 72 hours, fecal samples were recovered and dried for 48 hours Dried fecal samples were ground to a powdered from, and for each rat, 10 replicates of 50 mg samples were extracted with acetomtnle Insoluble mateπals were removed by centπfugation and supernatants analyzed by LC/MS/MS and compared against a standard curve to determine compound concentration The amount of compound actually dosed was determined by LC/MS/MS analysis of the dosing solutions The total amount of compound present in the 72 -hour fecal samples was compared to the total amount of compound dosed, and reported as percentage of total dose recovered The results, shown in Table 12, demonstrate near quantitative recovery of Examples 202 and 203 in 72-hour fecal samples
Table 12 Recover of dosed com ounds from 72-hour fecal sam les
Figure imgf000407_0001
All of the U S patents, U S patent application publications, U S patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification are incorporated herein by reference, in their entirety to the extent not inconsistent with the present descπption
From the foregoing it will be appreciated that, although specific embodiments of the invention have been descπbed herein for purposes of illustration, various modifications may be made without deviating from the spiπt and scope of the invention Accordingly, the invention is not limited except as by the appended claims

Claims

CLAIMSWhat is claimed is:
1. A compound having:
(i) a tPSA of at least about 200 A2 and a molecular weight of at least about 710 Daltons in the non-salt form; or
(ii) a tPSA of at least about 270 A2, wherein the compound is substantially active in the gastrointestinal tract to inhibit NHE-mediated antiport of sodium ions and hydrogen ions therein upon administration to a patient in need thereof.
2. A compound of claim 1 wherein the compound has a molecular weight of at least about 500 Da.
3. A compound of claim 1 or 2 wherein the compound has a molecular weight of at least about 1000 Da.
4. A compound of any of claims 1 -3 wherein the compound has a molecular weight of at least about 2500 Da.
5. A compound of any of claims 1-4 wherein the compound has a molecular weight of at least about about 5000 Da.
6. A compound of any of claims 1-5 wherein the compound has a tPSA of at least about 250 A2.
7. A compound of any of claims 1-6 wherein the compound has a tPSA of at least about 270 A2.
8. A compound of any of claims 1-7 wherein the compound has a tPSA of at least about 300 A2.
9. A compound of any of claims 1-8 wherein the compound has a tPSA of at least about 350 A2.
10. A compound of any of claims 1-9 wherein the compound has a tPSA of at least about 400 A2.
11. A compound of any of claims 1-10 wherein the compound has a tPSA of at least about 500 A2.
12. A compound of any of claims 1-11 wherein the compound is substantially active on the apical side of the epithelium of the gastrointestinal tract to inhibit antiport of sodium ions and hydrogen ions mediated by NHE-3, NHE-2, NHE-8, or a combination thereof.
13. A compound of any of claims 1-12 wherein the compound is substantially systemically non-bioavailable and/or substantially impermeable to the epithelium of the gastrointestinal tract.
14. A compound of any of claims 1-13 wherein the compound is substantially active in the lower gastrointestinal tract.
15. A compound of any of claims 1-14 wherein the compound has (i) a total number of NH and/or OH and/or other potential hydrogen bond donor moieties greater than about 5; (ii) a total number of O atoms and/or N atoms and/or other potential hydrogen bond acceptors greater than about 10; and/or (iii) a Moriguchi partition coefficient greater than about 105 or less than about 10.
16. A compound of any of claims 1-15 wherein the compound has a permeability coefficient, Papp, of less than about 100 x 10"6 cm/s, or less than about 1O x 10"6 cm/s, or less than about 1 x 10"6 cm/s, or less than about 0.1 x 10"6 cm/s.
17. A compound of any of claims 1-16 wherein the compound is substantially localized in the gastrointestinal tract or lumen.
18. A compound of any of claims 1-17 wherein the compound inhibits NHE irreversibly.
19. A compound of any of claims 1-18 wherein the compound is capable of providing a substantially persistent inhibitory action and wherein the compound is orally administered once-a-day.
20. A compound of any of claims 1-19 wherein the compound is substantially stable under physiological conditions in the gastrointestinal tract.
21. A compound of any of claims 1-20 wherein the compound is inert with regard to gastrointestinal flora.
22. A compound of any of claims 1-21 wherein the compound is designed to be delivered to the lower part of the gastrointestinal tract.
23. A compound of any of claims 1-22 wherein the compound is designed to be delivered to the lower part of the gastrointestinal tract past the duodenum.
24. A compound of any of claims 1-23 wherein the compound, when administered at a dose resulting in at least a 10% increase in fecal water content, has a Cmax that is less than the IC50 for NHE-3, less than about 1OX the IC50, or less than about IOOX the IC50.
25. A compound of any of claims 1-24 wherein, upon administration of the compound to a patient in need thereof, the compound exhibits a maximum concentration detected in the serum, defined as Cmax, that is lower than the NHE inhibitory concentration IC50 of the compound.
26. A compound of any of claims 1-24 wherein, upon administration of the compound to a patient in need thereof, greater than about 80%, greater than about 90% or greater than about 95% of the amount of compound administered is present in the patient's feces.
27. A compound of any of claims 1-26 wherein the compound has a structure of Formula (I) or (IX):
NHE — Z (I)
NHEH — Z
J E
(IX) wherein:
NHE is a NHE-inhibiting small molecule that comprises (i) a hetero- atom containing moiety, and (ii) a cyclic or heterocyclic scaffold or support moiety bound directly or indirectly thereto, the heteroatom-containing moiety being selected from a substituted guanidinyl moiety and a substituted heterocyclic moiety, which may optionally be fused with the scaffold or support moiety to form a fused bicyclic structure; and,
Z is a moiety having at least one site thereon for attachment to the NHE- inhibiting small molecule, the resulting NHE-Z molecule possessing overall physicochemical properties that render it substantially impermeable or substantially systemically non-bioavailable; and,
E is an integer having a value of 1 or more.
28. A compound of claim 27 wherein the total number of freely rotatable bonds in the NHE-Z molecule is at least about 10.
29. A compound of claim 27 or 28 wherein the total number hydrogen bond donors in the NHE-Z molecule is at least about 5.
30. A compound of any of claims 27-29 wherein the total number of hydrogen bond acceptors in the NHE-Z molecule is at least about 10.
31. A compound of any of claims 27-30 wherein the total number of hydrogen bond donors and hydrogen bond acceptors in the NHE-Z molecule is at least about 10.
32. A compound of any of claims 27-31 wherein the Log P of the NHE-Z inhibiting compound is at least about 5.
33. A compound of any of claims 27-31 wherein the log P of the NHE-Z inhibiting compound is less than about 1, or less than about 0.
34. A compound of any of claims 27-33 wherein the scaffold is a 5- member or 6-member cyclic or heterocyclic moiety.
35. A compound of claim 34 wherein the scaffold is aromatic.
36. A compound as set forth in any one of claims 27-35, wherein the scaffold of the NHE-inhibiting small molecule is bound to the moiety, Z, the compound having the structure of Formula (II): Substantially impermeable and/or substantially systemically non- bioavailablee NHE-inhibitinα compound f Λ
Figure imgf000413_0001
Y
NHE-inhibiting Small Molecule
(H) wherein:
Z is a Core having one or more sites thereon for attachment to one or more NHE-inhibiting small molecules, the resulting NHE-Z molecule possessing overall physicochemical properties that render it substantially impermeable or substantially systemically non-bioavailable;
B is the heteroatom-containing moiety of the NHE-inhibiting small molecule, and is selected from a substituted guanidinyl moiety and a substituted heterocyclic moiety, which may optionally be fused with the Scaffold moiety to form a fused, bicyclic structure;
Scaffold is the cyclic or heterocyclic scaffold or support moiety of the NHE-inhibiting small molecule, which is bound directly or indirectly to heteroatom- containing moiety, B, and which is optionally substituted with one or more additionally hydrocarbyl or heterohydrocarbyl moieties;
X is a bond or a spacer moiety selected from a group consisting of substituted or unsubstituted hydrocarbyl or heterohydrocarbyl moieties, and in particular substituted or unsubstituted Ci_7 hydrocarbyl or heterohydrocarbyl, and substituted or unsubstituted, saturated or unsaturated, cyclic or heterocyclic moieties, which links B and the Scaffold; and,
D and E are integers, each independently having a value of 1 or more.
37. A compound of any of claims 27-36 wherein the compound is an oligomer, dendrimer or polymer, and further wherein Z is a Core moiety having two or more sites thereon for attachment to multiple NHE-inhibiting small molecules, either directly or indirectly through a linking moiety, L, the compound having the structure of Formula (X):
Figure imgf000414_0001
(X)
wherein L is a bond or linker connecting the Core to the NHE-inhibiting small molecule, and n is an integer of 2 or more, and further wherein each NHE- inhibiting small molecule may be the same or differ from the others.
38. A compound of claim 37 wherein the NHE-inhibiting small molecule has the structure of Formula (IV):
Figure imgf000414_0002
(IV) or a stereoisomer, prodrug or pharmaceutically acceptable salt thereof, wherein: each R1, R2, R3, R5 and R9 are independently selected from H, halogen, - NR7(CO)R8, -(CO)NR7R8, -SO2-NR7R8, -NR7SO2R8, -NR7R8, -OR7, -SR7, - 0(CO)NR7R8, -NR7(CO)OR8, and -NR7SO2NR8, where R7 and R8 are independently selected from H or a bond linking the NHE-inhibiting small molecule to L, provided at least one is a bond linking the NHE-inhibiting small molecule to L;
R4 is selected from H, Ci-C7 alkyl, or a bond linking the NHE-inhibiting small molecule to L;
R6 is absent or selected from H and Cj-C7 alkyl; and ArI and Ar2 independently represent an aromatic ring or a heteroaromatic ring.
39. A compound of claim 38 wherein the NHE-inhibiting small molecule has the following structure:
Figure imgf000415_0001
or a stereoisomer, prodrug or pharmaceutically acceptable salt thereof, wherein: each R], R2 and R3 are independently selected from H, halogen, - NR7(CO)R8, -(CO)NR7R8, -SO2-NR7R8, -NR7SO2R8, -NR7R8, -OR7, -SR7, - 0(CO)NR7R8, -NR7(CO)OR8, and -NR7SO2NR8, where R7 and R8 are independently selected from H or a bond linking the NHE-inhibiting small molecule to L, provided at least one is a bond linking the NHE-inhibiting small molecule to L.
40. A compound of claim 39 wherein the NHE-inhibiting small molecule has one of the following structures:
Figure imgf000415_0002
or a stereoisomer, prodrug or pharmaceutically acceptable salt thereof.
41. A compound of any of claims 37-40 wherein L is a polyalkylene glycol linker.
42. A compound of any of claims 37-41 wherein L is a polyethylene glycol linker.
43. A compound of any of claims 37-42 wherein n is 2.
44. A compound of any of claims 37-43 wherein the Core has the following structure:
|-x-γ-x-|
wherein:
X is selected from the group consisting of a bond, -O-, -NH-, -S-, Ci- 6alkylene, -NHC(=O)-, -C(=O)NH-, -NHC(=O)NH-, -SO2NH-, and -NHSO2-;
Y is selected from the group consisting of a bond, optionally substituted Ci-8alkylene, optionally substituted aryl, optionally substituted heteroaryl, a polyethylene glycol linker, -(CH2) i_6O(CH2)1-6- and -(CH2)i_6NYi(CH2)i-6-; and
Y] is selected from the group consisting of hydrogen, optionally substituted Ci-salkyl, optionally substituted aryl or optionally substituted heteroaryl.
45. A compound of any of claims 37-44 wherein the Core is selected from the group consisting of:
Figure imgf000416_0001
Figure imgf000417_0001
46. A compound of any of claims 27-36 wherein the compound is an oligomer, and further wherein Z is a linking moiety, L, that links two or more NHE- inhibiting small molecules together, when the two or more NHE-inhibiting small molecules may be the same or different, the compound having the structure of Formula (XI):
NHEΞ-f— L NHEH — L NHE
\ ' m (XI)
wherein L is a bond or linker connecting one NHE-inhibiting small molecule to another, and m is 0 or an integer of 1 or more.
47. A compound of any of claims 27-36 wherein the compound is an oligomer, dendrimer or polymer, and further wherein Z is a backbone, denoted Repeat Unit, to which is bound multiple NHE-inhibiting moieties, the compound having the structure of Formula (XIIB):
— I — I repeat
Figure imgf000418_0001
(XIIB)
wherein: L is a bond or a linking moiety; NHE is a NHE-inhibiting small molecule; and n is a non-zero integer.
48. A pharmaceutical composition comprising a compound of any of claims 1 -47, or a stereoisomer, pharmaceutically acceptable salt or prodrug thereof, and a pharmaceutically acceptable carrier, diluent or excipient.
49. A pharmaceutical composition of claim 48, further comprising a fluid-absorbing polymer.
50. A pharmaceutical composition of claim 49 wherein the fluid- absorbing polymer is delivered directly to the colon.
51. A pharmaceutical composition of claim 49 or 50 wherein the fluid-absorbing polymer has a fluid absorbency of at least about 15 g of isotonic fluid per g of polymer under a static pressure of about 5 kPa.
52. A pharmaceutical composition of any of claims 49-51 wherein the fluid-absorbing polymer has a fluid absorbency of at least about 15 g of isotonic fluid per g of polymer under a static pressure of about 10 kPa.
53. A pharmaceutical composition of any of claims 49-52 wherein the fluid-absorbing polymer is characterized by a fluid absorbency of at least about 10 g/g-
54. A pharmaceutical composition of any of claims 49-53 wherein the fluid-absorbing polymer is characterized by a fluid absorbency of at least about 15 g/g-
55. A pharmaceutical composition of any of claims 49-54 wherein the fluid-absorbing polymer is superabsorbent.
56. A pharmaceutical composition of any of claims 49-54 wherein the fluid-absorbing polymer is a crosslinked, partially neutralized polyelectrolyte hydrogel.
57. A pharmaceutical composition of any of claims 49-54 wherein the fluid-absorbing polymer is a crosslinked polyacrylate.
58. A pharmaceutical composition of any of claims 49-54 wherein the fluid-absorbing polymer is a polyelectrolyte.
59. A pharmaceutical composition of any of claims 49-54 wherein the fluid-absorbing polymer is calcium Carbophil.
60. A pharmaceutical composition of any of claims 49-54 wherein the fluid-absorbing polymer is prepared by a high internal phase emulsion process.
61. A pharmaceutical composition of any of claims 49-54 wherein the fluid-absorbing polymer is a foam.
62. A pharmaceutical composition of any of claims 49-54 wherein the fluid-absorbing polymer is prepared by a aqueous free radical polymerization of acrylamide or a derivative thereof, a crosslinker and a free radical initiator redox system in water.
63. A pharmaceutical composition of any of claims 49-54 wherein the fluid-absorbing polymer is a hydrogel.
64. A pharmaceutical composition of any of claims 49-54 wherein the fluid-absorbing polymer is an N-alkyl acrylamide.
65. A pharmaceutical composition of any of claims 49-54 wherein the fluid-absorbing polymer is a superporous gel.
66. A pharmaceutical composition of any of claims 49-54 wherein the fluid-absorbing polymer is naturally occurring.
67. A pharmaceutical composition of any of claims 49-54 wherein the fluid-absorbing polymer is selected from the group consisting of xanthan, guar, wellan, hemicelluloses, alkyl-cellulose hydro-alkyl-cellulose, carboxy-alkyl-cellulose, carrageenan, dextran, hyaluronic acid and agarose.
68. A pharmaceutical composition of any of claims 49-54 wherein the fluid-absorbing polymer is psyllium.
69. A pharmaceutical composition of any of claims 49-54 wherein the fluid-absorbing polymer is a polysaccharide that includes xylose and arabinose.
70. A pharmaceutical composition of any of claims 49-54 wherein the fluid-absorbing polymer is a polysaccharide that includes xylose and arabinose, wherein the ratio of xylose to arabinose is at least about 3:1, by weight.
71. A pharmaceutical composition of any of claims 49-70, further comprising another pharmaceutically active agent or compound.
72. A pharmaceutical composition of claim 71 wherein the composition further comprises another pharmaceutically active agent or compound selected from the group consisting of a diuretic, cardiac glycoside, ACE inhibitor, angiotensin-2 receptor antagonist, calcium channel blocker, beta blocker, alpha blocker, central alpha agonist, vasodilator, blood thinner, anti-platelet agent, lipid-lowering agent, and peroxisome proliferator-activated receptor (PPAR) gamma agonist agent.
73. A pharmaceutical composition of claim 72 wherein the diuretic is selected from the group consisting of a high ceiling loop diuretic, a benzothiadiazide diuretic, a potassium sparing diuretic, and a osmotic diuretic.
74. A pharmaceutical composition of claim 71 wherein the composition further comprises another pharmaceutically active agent or compound selected from the group consisting of an analgesic peptide or agent.
75. A pharmaceutical composition of claim 74 wherein the composition further comprises another pharmaceutically active agent or compound selected from the group consisting of a laxative agent selected from a bulk-producing agent (e.g. psyllium husk (Metamucil)), methylcellulose (Citrucel), polycarbophil, dietary fiber, apples, stool softeners/surfactant (e.g., docusate, Colace, Diocto), a hydrating or osmotic agent (e.g., dibasic sodium phosphate, magnesium citrate, magnesium hydroxide (Milk of magnesia), magnesium sulfate (which is Epsom salt), monobasic sodium phosphate, sodium biphosphate), a hyperosmotic agent (e.g., glycerin suppositories, sorbitol, lactulose, and polyethylene glycol (PEG)).
76. A method for inhibiting NHE-mediated antiport of sodium and hydrogen ions, the method comprising administering to a mammal in need thereof a pharmaceutically effective amount of a compound or pharmaceutical composition of any of claims 1-75.
77. A method for treating a disorder associated with fluid retention or salt overload, the method comprising administering to a mammal in need thereof a pharmaceutically effective amount of a compound or pharmaceutical composition of any of claims 1-75.
78. A method for treating a disorder selected from the group consisting of heart failure, chronic kidney disease, end-stage renal disease, liver disease, and peroxisome proliferator-activated receptor (PPAR) gamma agonist-induced fluid retention, the method comprising administering to a mammal in need thereof a pharmaceutically effective amount of a compound or pharmaceutical composition of any of claims 1-75.
79. The method of claim 78 wherein the heart failure is congestive heart failure.
80. A method for treating hypertension, the method comprising administering to a mammal in need thereof a pharmaceutically effective amount of a compound or pharmaceutical composition of any of claims 1-75.
81. A method of any of claims 77-80 wherein the method comprises administering a pharmaceutically effective amount of the compound to the mammal in order to increase the mammal's daily fecal output of sodium and/or fluid.
82. A method of any of claims 77-81 wherein the method comprises administering a pharmaceutically effective amount of the compound to the mammal in order to increase the mammal's daily fecal output of sodium by at least about 30 mmol, and/or fluid by at least about 200 ml.
83. A method of any of claims 77-82 wherein the mammal's fecal output of sodium and/or fluid is increased without introducing another type of cation in a stoichiometric or near stoichiometric fashion via an ion exchange process.
84. A method of any of claims 77-83, further comprising administering to the mammal a fluid-absorbing polymer to absorb fecal fluid resulting from the use of the compound that is substantially active in the gastrointestinal tract to inhibit NHE-mediated antiport of sodium ions and hydrogen ions therein.
85. A method of any of claims 77-84 wherein the compound or composition is administered to treat hypertension.
86. A method of any of claims 77-85 wherein the compound or composition is administered to treat hypertension associated with dietary salt intake.
87. A method of any of claims 77-84 wherein administration of the compound or composition allows the mammal to intake a more palatable diet.
88. A method of any of claims 77-84 wherein the compound or composition is administered to treat fluid overload.
89. A method of claim 88 wherein the fluid overload is associated with congestive heart failure.
90. A method of claim 88 wherein the fluid overload is associated with end stage renal disease.
91. A method of claim 88 wherein the fluid overload is associated with peroxisome proliferator-activated receptor (PPAR) gamma agonist therapy.
92. A method of any of claims 77-84 wherein the compound or composition is administered to treat sodium overload.
93. A method of any of claim 77-84 wherein the compound or composition is administered to reduce interdialytic weight gain in ESRD patients.
94. A method of any of claims 77-84 wherein the compound or composition is administered to treat edema.
95. A method of any of claims 94 wherein the edema is caused by chemotherapy, pre-menstrual fluid overload or preeclampsia.
96. A method of any of claims 77-95 wherein the compound or composition is administered orally, by rectal suppository, or enema.
97. A method of any one of claims 77-96, wherein the method comprises administering a pharmaceutically effective amount of the compound or composition in combination with one or more additional pharmaceutically active compounds or agents.
98. A method of claim 97 wherein the one or more additional pharmaceutically active compounds or agents is selected from the group consisting of a diuretic, cardiac glycoside, ACE inhibitor, angiotensin-2 receptor antagonist, aldosterone antagonist, calcium channel blocker, beta blocker, alpha blocker, central alpha agonist, vasodilator, blood thinner, anti-platelet agent, lipid-lowering agent, and peroxisome proliferator-activated receptor (PPAR) gamma agonist agent.
99. A method of claim 98 wherein the diuretic is selected from the group consisting of a high ceiling loop diuretic, a benzothiadiazide diuretic, a potassium sparing diuretic, and a osmotic diuretic.
100. A method of any of claims 97-99 wherein the pharmaceutically effective amount of the compound or composition, and the one or more additional pharmaceutically active compounds or agents, are administered as part of a single pharmaceutical preparation.
101. A method of any of claims 97-99 wherein the pharmaceutically effective amount of the compound or composition, and the one or more additional pharmaceutically active compounds or agents, are administered as individual pharmaceutical preparations.
102. A method of claim 101 wherein the individual pharmaceutical preparation are administered sequentially.
103. A method of claim 102 wherein the individual pharmaceutical preparation are administered simultaneously.
104. A method for treating a gastrointestinal tract disorder, the method comprising administering to a mammal in need thereof a pharmaceutically effective amount of a compound or pharmaceutical composition of any of claims 1-75.
105. A method of claim 104 wherein the gastrointestinal tract disorder is a gastrointestinal motility disorder.
106. A method of claim 104 wherein the gastrointestinal tract disorder is irritable bowel syndrome.
107. A method of claim 104 wherein the gastrointestinal tract disorder is chronic constipation.
108. A method of claim 104 wherein the gastrointestinal tract disorder is chronic idiopathic constipation.
109. A method of claim 104 wherein the gastrointestinal tract disorder is chronic constipation occurring in cystic fibrosis patients.
110. A method of claim 104 wherein the gastrointestinal tract disorder is opioid-induced constipation.
111. A method of claim 104 wherein the gastrointestinal tract disorder is a functional gastrointestinal tract disorder.
112. A method of claim 104 wherein the gastrointestinal tract disorder is selected from the group consisting of chronic intestinal pseudo-obstruction and colonic pseudo-obstruction.
113. A method of claim 104 wherein the gastrointestinal tract disorder is Crohn's disease.
114. A method of claim 104 wherein the gastrointestinal tract disorder is ulcerative colitis.
115. A method of claim 104 wherein the gastrointestinal tract disorder is a disease referred to as inflammatory bowel disease.
116. A method of claim 104 wherein the gastrointestinal tract disorder is associated with chronic kidney disease (stage 4 or 5).
117. A method of claim 104 wherein the gastrointestinal tract disorder is constipation induced by calcium supplement.
118. A method of claim 104 wherein the gastrointestinal tract disorder is constipation, and further wherein the constipation to be treated is associated with the use of a therapeutic agent.
119. A method of claim 104 wherein the gastrointestinal tract disorder is constipation, and further wherein the constipation to be treated is associated with a neuropathic disorder.
120. A method of claim 104 wherein the gastrointestinal tract disorder is constipation, and further wherein the constipation to be treated is post-surgical constipation (postoperative ileus).
121. A method of claim 104 wherein the gastrointestinal tract disorder is constipation, and further wherein the constipation to be treated is idiopathic (functional constipation or slow transit constipation).
122. A method of claim 104 wherein the gastrointestinal tract disorder is constipation, and further wherein the constipation to be treated is associated with neuropathic, metabolic or an endocrine disorder (e.g., diabetes mellitus, renal failure, hypothyroidism, hyperthyroidism, hypocalcaemia, Multiple Sclerosis, Parkinson's disease, spinal cord lesions, neurofibromatosis, autonomic neuropathy, Chagas disease, Hirschsprung's disease or cystic fibrosis, and the like).
123. A method of claim 104 wherein the gastrointestinal tract disorder is constipation, and further wherein the constipation to be treated is due the use of drugs selected from analgesics (e.g., opioids), antihypertensives, anticonvulsants, antidepressants, antispasmodics and antipsychotics.
124. A method for treating irritable bowel syndrome, the method comprising administering to a mammal in need thereof a pharmaceutically effective amount of an NHE-3 inhibitor compound or a pharmaceutical composition comprising an NHE-3 inhibitor compound.
125. A method of claim 124 wherein the NHE-3 inhibitor compound or the pharmaceutical composition comprising an NHE-3 inhibitor compound is a compound or pharmaceutical composition of any of claims 1-75.
126. A method of any of claims 104-125 wherein the compound or composition is administered to treat or reduce pain associated with a gastrointestinal tract disorder.
127. A method of any of claims 104-125 wherein the compound or composition is administered to treat or reduce visceral hypersensitivity associated with a gastrointestinal tract disorder.
128. A method of any of claims 104-125 wherein the compound or composition is administered to treat or reduce inflammation of the gastrointestinal tract.
129. A method of any of claims 104-125 wherein the compound or composition is administered to reduce gastrointestinal transit time.
130. A method of any of claims 104-129 wherein the compound or composition is administered either orally or by rectal suppository.
131. A method of any of claims 104-130 wherein the method comprises administering a pharmaceutically effective amount of the compound or composition, in combination with one or more additional pharmaceutically active compounds or agents.
132. A method of claim 131 wherein the one or more additional pharmaceutically active agents or compounds are an analgesic peptide or agent.
133. A method of claim 131 wherein the one or more additional pharmaceutically active agents or compounds are selected from the group consisting of a laxative agent selected from a bulk-producing agent (e.g. psyllium husk (Metamucil)), methylcellulose (Citrucel), polycarbophil, dietary fiber, apples, stool softeners/surfactant (e.g., docusate, Colace, Diocto), a hydrating or osmotic agent (e.g., dibasic sodium phosphate, magnesium citrate, magnesium hydroxide (Milk of magnesia), magnesium sulfate (which is Epsom salt), monobasic sodium phosphate, sodium biphosphate), and a hyperosmotic agent (e.g., glycerin suppositories, sorbitol, lactulose, and polyethylene glycol (PEG)).
134. A method of any of claims 131-133 wherein the pharmaceutically effective amount of the compound or composition, and the one or more additional pharmaceutically active compounds or agents, are administered as part of a single pharmaceutical preparation.
135. A method of any of claims 131-133 wherein the pharmaceutically effective amount of the compound or composition, and the one or more additional pharmaceutically active compounds or agents, are administered as individual pharmaceutical preparations.
136. A method of claim 135 wherein the individual pharmaceutical preparation are administered sequentially.
137. A method of claim 135 wherein the individual pharmaceutical preparation are administered simultaneously.
PCT/US2009/069852 2008-12-31 2009-12-30 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders WO2010078449A2 (en)

Priority Applications (34)

Application Number Priority Date Filing Date Title
EP09796924.0A EP2384318B1 (en) 2008-12-31 2009-12-30 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
JP2011543730A JP5502106B2 (en) 2008-12-31 2009-12-30 Compositions and methods for inhibiting NHE-mediated antiports in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal disorders
PL09796924T PL2384318T3 (en) 2008-12-31 2009-12-30 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
EP17200784.1A EP3351248B1 (en) 2008-12-31 2009-12-30 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
ES09796924.0T ES2657938T3 (en) 2008-12-31 2009-12-30 Compounds and methods to inhibit NHE-mediated anti-port in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
KR1020177021507A KR20170091783A (en) 2008-12-31 2009-12-30 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
MX2011007024A MX2011007024A (en) 2008-12-31 2009-12-30 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders.
BRPI0923861A BRPI0923861B8 (en) 2008-12-31 2009-12-30 compounds and pharmaceutical compositions for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
MX2015004407A MX345283B (en) 2008-12-31 2009-12-30 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders.
KR1020227009722A KR20220042487A (en) 2008-12-31 2009-12-30 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
DK09796924.0T DK2384318T3 (en) 2008-12-31 2009-12-30 RELATIONS AND PROCEDURES FOR INHIBITING NHE-MEDIATED ANTI-PORTION FOR TREATING DISEASES ASSOCIATED WITH FLUID RETENTION OR SALT LOADING AND DISEASE DISEASES
KR1020167033454A KR101766619B1 (en) 2008-12-31 2009-12-30 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
KR1020117017941A KR101683318B1 (en) 2008-12-31 2009-12-30 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
AU2009334511A AU2009334511C1 (en) 2008-12-31 2009-12-30 Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
CN2009801576148A CN102333759A (en) 2008-12-31 2009-12-30 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
CA2748607A CA2748607A1 (en) 2008-12-31 2009-12-30 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
SI200931806T SI2384318T1 (en) 2008-12-31 2009-12-30 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
EP21178147.1A EP3939964A1 (en) 2008-12-31 2009-12-30 Combinations for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
LTEP09796924.0T LT2384318T (en) 2008-12-31 2009-12-30 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
NO09796924A NO2384318T3 (en) 2008-12-31 2009-12-30
KR1020207024357A KR20200111230A (en) 2008-12-31 2009-12-30 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
US13/172,394 US8541448B2 (en) 2008-12-31 2011-06-29 Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
IL213852A IL213852A (en) 2008-12-31 2011-06-30 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
US13/826,186 US9006281B2 (en) 2008-12-31 2013-03-14 Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
US13/804,752 US8969377B2 (en) 2008-12-31 2013-03-14 Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
US14/592,200 US9408840B2 (en) 2008-12-31 2015-01-08 Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorder
US15/402,211 US10543207B2 (en) 2008-12-31 2017-01-09 Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
IL250641A IL250641B (en) 2008-12-31 2017-02-16 Pharmaceutical compositions for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
US16/476,838 US20190374533A1 (en) 2008-12-31 2018-01-09 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
CY20181100147T CY1120451T1 (en) 2008-12-31 2018-02-06 COMPOUNDS AND METHODS FOR SUSPENDED MEDIUM SUSPENSION WITH INTERFERENCE IN DISORDER TREATMENT RELATED TO DETERMINATION OF EFFECTS
HRP20180289TT HRP20180289T1 (en) 2008-12-31 2018-02-15 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorderss
IL259851A IL259851B (en) 2008-12-31 2018-06-06 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
US16/773,723 US11318129B2 (en) 2008-12-31 2020-01-27 Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
US17/711,863 US20230031776A1 (en) 2008-12-31 2022-04-01 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US14185308P 2008-12-31 2008-12-31
US61/141,853 2008-12-31
US16950909P 2009-04-15 2009-04-15
US61/169,509 2009-04-15
US23784209P 2009-08-28 2009-08-28
US61/237,842 2009-08-28

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/172,394 Continuation US8541448B2 (en) 2008-12-31 2011-06-29 Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders

Publications (2)

Publication Number Publication Date
WO2010078449A2 true WO2010078449A2 (en) 2010-07-08
WO2010078449A3 WO2010078449A3 (en) 2010-09-30

Family

ID=41650053

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/069852 WO2010078449A2 (en) 2008-12-31 2009-12-30 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders

Country Status (22)

Country Link
US (4) US8541448B2 (en)
EP (3) EP3939964A1 (en)
JP (2) JP5502106B2 (en)
KR (5) KR20170091783A (en)
CN (2) CN102333759A (en)
AU (1) AU2009334511C1 (en)
BR (1) BRPI0923861B8 (en)
CA (1) CA2748607A1 (en)
CY (1) CY1120451T1 (en)
DK (1) DK2384318T3 (en)
ES (1) ES2657938T3 (en)
HK (1) HK1198162A1 (en)
HR (1) HRP20180289T1 (en)
HU (1) HUE036405T2 (en)
IL (3) IL213852A (en)
LT (1) LT2384318T (en)
MX (2) MX2011007024A (en)
NO (1) NO2384318T3 (en)
PL (1) PL2384318T3 (en)
PT (1) PT2384318T (en)
SI (1) SI2384318T1 (en)
WO (1) WO2010078449A2 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102757390A (en) * 2011-04-27 2012-10-31 中国中化股份有限公司 Method for preparing 2-methoxy-4-diazanyl-5-fluoropyrimidine
WO2014029984A1 (en) * 2012-08-21 2014-02-27 Ardelyx, Inc. Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
WO2014029983A1 (en) 2012-08-21 2014-02-27 Ardelyx, Inc. Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
KR20170034897A (en) 2014-07-25 2017-03-29 다이쇼 세이야꾸 가부시끼가이샤 Phenyl tetrahydroisoquinoline compound substituted with heteroaryl
US20170368004A1 (en) * 2012-08-21 2017-12-28 Ardelyx, Inc Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
WO2018129552A1 (en) 2017-01-09 2018-07-12 Ardelyx, Inc. Compounds useful for treating gastrointestinal tract disorders
WO2018129557A1 (en) 2017-01-09 2018-07-12 Ardelyx, Inc. Inhibitors of nhe-mediated antiport
WO2018129556A1 (en) * 2017-01-09 2018-07-12 Ardelyx, Inc. Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
WO2019060051A1 (en) 2017-08-04 2019-03-28 Ardelyx, Inc. Glycyrrhetinic acid derivatives for treating hyperkalemia
US10272079B2 (en) 2013-04-12 2019-04-30 Ardelyx, Inc. NHE3-binding compounds and methods for inhibiting phosphate transport
WO2019091503A1 (en) 2017-11-13 2019-05-16 Zentiva K.S. Solid forms of tenapanor and method of preparation of tenapanor
US10543207B2 (en) 2008-12-31 2020-01-28 Ardelyx, Inc. Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
WO2020051014A1 (en) 2018-09-04 2020-03-12 Teva Pharmaceuticals International Gmbh Processes for the preparation of tenapanor and intermediates thereof
WO2020163642A1 (en) 2019-02-07 2020-08-13 Ardelyx, Inc. Glycyrrhetinic acid derivatives for use in treating hyperkalemia
WO2020231770A1 (en) 2019-05-16 2020-11-19 Eli Lilly And Company Sodium-hydrogen exchanger 3 inhibitor compounds
WO2020237096A1 (en) 2019-05-21 2020-11-26 Ardelyx, Inc. Combination for lowering serum phosphate in a patient
CN112047881A (en) * 2020-10-10 2020-12-08 山东汇海医药化工有限公司 Preparation method of tanaprost
CN112679427A (en) * 2019-10-17 2021-04-20 江苏恒瑞医药股份有限公司 Tetrahydroisoquinoline derivatives, preparation method and medical application thereof
US11938134B2 (en) 2017-03-10 2024-03-26 Eikonizo Therapeutics, Inc. Metalloenzyme inhibitor compounds

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20170091783A (en) 2008-12-31 2017-08-09 알데릭스, 인코포레이티드 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
EP3016944A4 (en) 2013-07-02 2016-11-16 California Inst Biomedical Res Compounds for treatment of cystic fibrosis
MX2016001714A (en) * 2013-08-09 2016-10-03 Ardelyx Inc Compounds and methods for inhibiting phosphate transport.
WO2015084158A1 (en) * 2013-12-06 2015-06-11 N.V. Nutricia A pyrimidine derivative and a fatty acid source for use in the treatment of constipation
DE102014019388A1 (en) 2014-12-29 2016-06-30 Susanne Wagner Maghemite-based medicines for the simultaneous reduction of gastrointestinal sodium absorption and phosphate absorption
CN107405505A (en) * 2015-01-07 2017-11-28 加州生物医学研究所 For treating the compound of cystic fibrosis
JP6421100B2 (en) * 2015-09-04 2018-11-07 株式会社ダイセル Method for producing carboxamide compound
US10016379B2 (en) * 2015-10-30 2018-07-10 Robin L. Webb Method of treatment for third spacing
BR112020008676A2 (en) * 2017-11-02 2020-10-27 Ube Industries, Ltd. compound, and pharmaceutical composition.
CN109897044A (en) * 2017-12-07 2019-06-18 北京大学 Anti- neurogenic pain compound and preparation method thereof
CN109321529A (en) * 2018-10-19 2019-02-12 浙江工商大学 A kind of construction method of external gastrointestinal model and application
CN111954669B (en) * 2019-03-14 2023-04-07 深圳仁泰医药科技有限公司 Crystal form A of NHE3 inhibitor, and preparation method and application thereof
AU2021401102B2 (en) * 2020-12-18 2024-03-07 Jiangxi Jemincare Group Co., Ltd Benzoheterocycle substituted tetrahydroisoquinoline compound
WO2023246732A1 (en) * 2022-06-20 2023-12-28 上海济煜医药科技有限公司 Benzoheterocyclic substituted tetrahydroisoquinoline compound salt form and preparation method therefor

Family Cites Families (130)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3926891A (en) 1974-03-13 1975-12-16 Dow Chemical Co Method for making a crosslinkable aqueous solution which is useful to form soft, water-swellable polyacrylate articles
US3935099A (en) 1974-04-03 1976-01-27 The United States Of America As Represented By The Secretary Of Agriculture Method of reducing water content of emulsions, suspensions, and dispersions with highly absorbent starch-containing polymeric compositions
US3997484A (en) 1974-04-03 1976-12-14 The United States Of America As Represented By The Secretary Of Agriculture Highly-absorbent starch-containing polymeric compositions
US4090013A (en) 1975-03-07 1978-05-16 National Starch And Chemical Corp. Absorbent composition of matter
JPS51125468A (en) 1975-03-27 1976-11-01 Sanyo Chem Ind Ltd Method of preparing resins of high water absorbency
JPS5346389A (en) 1976-10-07 1978-04-25 Kao Corp Preparation of self-crosslinking polymer of acrylic alkali metal salt
US4190562A (en) 1977-04-04 1980-02-26 The B. F. Goodrich Company Improved water absorbent copolymers of copolymerizable carboxylic acids and acrylic or methacrylic esters
US4470975A (en) 1977-10-21 1984-09-11 The Johns Hopkins University Method and composition for the elimination of water from an animal body
US4286082A (en) 1979-04-06 1981-08-25 Nippon Shokubai Kagaku Kogyo & Co., Ltd. Absorbent resin composition and process for producing same
JPS6025045B2 (en) 1980-03-19 1985-06-15 製鉄化学工業株式会社 Method for producing acrylic acid polymer with excellent salt water absorption ability
JPS57158209A (en) 1981-03-25 1982-09-30 Kao Corp Production of bead-form highly water-absorbing polymer
JPS5832607A (en) 1981-08-20 1983-02-25 Kao Corp Preparation of water-absorbing material having improved water absorption property
US4985518A (en) 1981-10-26 1991-01-15 American Colloid Company Process for preparing water-absorbing resins
JPS6187702A (en) 1984-10-05 1986-05-06 Seitetsu Kagaku Co Ltd Production of water-absorptive resin
US4708997A (en) 1985-07-22 1987-11-24 The Dow Chemical Company Suspending agent for the suspension polymerization of water-soluble monomers
US4806532A (en) 1985-10-08 1989-02-21 Mayo Foundation For Medical Education And Research Inhibition of epithelial phosphate transport
DE3544770A1 (en) 1985-12-18 1987-06-19 Stockhausen Chem Fab Gmbh METHOD AND DEVICE FOR THE CONTINUOUS PRODUCTION OF POLYMERISATES AND COPOLYMERISATES OF ACRYLIC ACID AND / OR METHACRYLIC ACID
US4766004A (en) 1986-12-19 1988-08-23 Warner-Lambert Company Crunchy, highly palatable, bulk-increasing, dietary fiber supplement composition
IT1217123B (en) 1987-02-05 1990-03-14 Rotta Research Lab OPTICALLY ACTIVE DERIVATIVES OF ACID 5 PENTILAMINE 5 OXO PENTANOIC R WITH ANTAGONIST ACTIVITY OF THE CHOLECISTOKININ AND PROCEDURE FOR THEIR PREPARATION
US4999200A (en) 1987-12-09 1991-03-12 Marion Laboratories Psyllium tablet composition, method of manufacture and method of use
US5149541A (en) 1988-10-03 1992-09-22 The Procter & Gamble Company Psyllium-containing produces with a distribution of particle size
US5145906A (en) 1989-09-28 1992-09-08 Hoechst Celanese Corporation Super-absorbent polymer having improved absorbency properties
US5126150A (en) 1990-10-01 1992-06-30 The Procter & Gamble Company Compositions containing psyllium
HUT64023A (en) 1991-03-22 1993-11-29 Sandoz Ag Process for producing aminoguanidine derivatives and pharmaceutical compositions comprising such compounds
FR2674849B1 (en) 1991-04-02 1994-12-23 Logeais Labor Jacques NOVEL N-CYCLOHEXYL BENZAMIDES OR THIOBENZAMIDES DERIVATIVES, THEIR PREPARATIONS AND THEIR THERAPEUTIC APPLICATIONS.
US5140102A (en) 1991-09-23 1992-08-18 Monsanto Company Pentadecapeptide, guanylin, which stimulates intestinal guanylate cyclase
US5824645A (en) 1991-12-30 1998-10-20 Neurex Corporation Method of treating inflammation
EP0625162B1 (en) 1991-12-30 1998-05-13 Neurex Corporation Methods of producing analgesia and enhancing opiate analgesia
US5969097A (en) 1992-06-23 1999-10-19 G. D. Searle & Co. Human guanylin
ATE157351T1 (en) 1993-02-20 1997-09-15 Hoechst Ag SUBSTITUTED BENZOYLGUANIDINES, METHOD FOR THE PRODUCTION THEREOF, THEIR USE AS A MEDICATION, AS INHIBITORS OF CELLULAR NA+/H+ EXCHANGE OR AS A DIAGNOSTIC, AND MEDICINE CONTAINING THEM
MX213505B (en) 1993-02-24 2003-04-03
US5629377A (en) 1993-03-10 1997-05-13 The Dow Chemical Company Water absorbent resin particles of crosslinked carboxyl containing polymers and method of preparation
IL109570A0 (en) 1993-05-17 1994-08-26 Fujisawa Pharmaceutical Co Guanidine derivatives, pharmaceutical compositions containing the same and processes for the preparation thereof
US5489670A (en) 1993-10-29 1996-02-06 G. D. Searle & Co. Human uroguanylin
US5610145A (en) 1994-04-15 1997-03-11 Warner-Lambert Company Tachykinin antagonists
US5650222A (en) 1995-01-10 1997-07-22 The Procter & Gamble Company Absorbent foam materials for aqueous fluids made from high internal phase emulsions having very high water-to-oil ratios
DE19518796A1 (en) 1995-05-22 1996-11-28 Hoechst Ag Fluorophenyl-substituted alkenylcarboxylic acid guanidines, processes for their preparation, their use as medicaments or diagnostic agents and medicaments containing them
US6054429A (en) 1996-03-08 2000-04-25 Elan Pharmaceuticals, Inc. Epidural method of producing analgesia
US5795864A (en) 1995-06-27 1998-08-18 Neurex Corporation Stable omega conopetide formulations
DK0835126T3 (en) 1995-06-27 2003-07-14 Elan Pharm Inc Compositions and Formulations for the Preparation of Analgesic Drugs and for Inhibition of Progressive Neuropathic Amniotic Disorders
US5550167A (en) 1995-08-30 1996-08-27 The Procter & Gamble Company Absorbent foams made from high internal phase emulsions useful for acquiring aqueous fluids
AP9801237A0 (en) 1995-11-24 1998-06-30 Smithkline Beecham Farm S P A Quinoline derivatives.
GB9524104D0 (en) 1995-11-24 1996-01-24 Smithkline Beecham Spa Novel compounds
DE19548812A1 (en) 1995-12-27 1997-07-03 Hoechst Ag Use of inhibitors of the cellular Na · + · / H · + · exchanger (NHE) for the manufacture of a medicament for respiratory stimulation
EP0837055A1 (en) 1996-07-30 1998-04-22 Hoechst Aktiengesellschaft Substituted Indanylidineacetylguanidines, process for their preparation, their use as medicaments or diagnostic and medicaments containing them
DE19633966A1 (en) 1996-08-22 1998-02-26 Hoechst Ag Phenyl-substituted alkenylcarboxylic acid guanidines, process for their preparation, their use as a medicament or diagnostic agent, and medicament containing them
AU4176997A (en) 1996-09-16 1998-04-02 Warner-Lambert Company 3-alkyl-3-phenyl-piperidines
DK0975347T3 (en) 1997-02-28 2008-06-16 Nycomed Gmbh Synergistic combination of PDE inhibitors and adenylate cyclase agonists or guanyl cyclist agonists
NZ329807A (en) 1997-04-23 2000-07-28 Pfizer NK-1 receptor antagonists and P receptor antagonists 2-Diarylmethyl-3-amino-1-azabicyclo[2.2.2]octane derivatives and amino substituted N-containing rings as agents for treating irritable bowel syndrome
KR100219918B1 (en) 1997-07-03 1999-09-01 김윤 Composition for colon specific drug delivery
US20010006972A1 (en) 1998-04-21 2001-07-05 Stephen A. Williams Nk-1 receptor antagonists for the treatment of symptoms of irritable bowel syndrome
KR20000011247A (en) 1998-07-23 2000-02-25 김윤 Composition and pharmaceutical dosage form for colonic drug delivery using polysaccharides
DE19849722A1 (en) 1998-10-28 2000-05-04 Aventis Pharma Gmbh Substituted phenyl-alkenoylguanidines, processes for their preparation, their use as medicaments or diagnostic agents and medicaments containing them
US6355823B1 (en) 1999-01-21 2002-03-12 Board Of Regents, The University Of Texas System Inhibitors of intestinal apical membrane na/phoshate co-transportation
US6624150B2 (en) 1999-02-26 2003-09-23 Inspire Pharmaceuticals, Inc. Method of treating gastrointestinal tract disease with purinergic receptor agonists
US6287609B1 (en) 1999-06-09 2001-09-11 Wisconsin Alumni Research Foundation Unfermented gel fraction from psyllium seed husks
US6475509B1 (en) 1999-07-19 2002-11-05 The University Of North Carolina At Chapel Hill Covalent conjugates of sodium channel blockers and active compounds
AR030911A1 (en) 1999-07-20 2003-09-03 Smithkline Beecham Corp USE OF N-ARIL-2-SULFONAMIDOBENZAMIDS FOR THE MANUFACTURE OF A MEDICINAL PRODUCT FOR THE TREATMENT OF CHRONIC RENAL FAILURE AND PHARMACEUTICAL COMPOSITIONS
US6107356A (en) 1999-08-23 2000-08-22 The Procter & Gamble Company High suction polymeric foam materials
DE19941764A1 (en) * 1999-09-02 2001-03-15 Aventis Pharma Gmbh Substituted acylguanidines, processes for their preparation, their use as medicines or diagnostics and medicines containing them
SE9903291D0 (en) 1999-09-15 1999-09-15 Astra Ab New process
DE19945302A1 (en) 1999-09-22 2001-03-29 Merck Patent Gmbh Biphenyl derivatives as NHE-3 inhibitors
US6887870B1 (en) 1999-10-12 2005-05-03 Bristol-Myers Squibb Company Heterocyclic sodium/proton exchange inhibitors and method
ES2226933T3 (en) 1999-11-01 2005-04-01 John Rhodes COMPOSITION TO TREAT STRETCHING AND IRRITABLE INTESTINE SYNDROME.
AUPQ514600A0 (en) 2000-01-18 2000-02-10 James Cook University Brain injury treatment
WO2001064212A1 (en) 2000-03-01 2001-09-07 University College London Modulators of the endocannabinoid uptake and of the vallinoid receptors
DE10015248A1 (en) 2000-03-28 2001-10-04 Merck Patent Gmbh Bisamidino compounds as NHE-3 inhibitors
DE10019062A1 (en) 2000-04-18 2001-10-25 Merck Patent Gmbh Use of known and new 2-guanidino-4-aryl-quinazoline derivatives as NHE-3 inhibitors useful for the treatment of e.g. hypertension, thrombosis, cardiac ischemia, peripheral and CNS ischemia
AU2001259354A1 (en) 2000-05-02 2001-11-12 Smith Kline Beecham Corporation Phosphate transport inhibitors
JP2003533477A (en) 2000-05-12 2003-11-11 スミスクライン・ビーチャム・コーポレイション Phosphate transport inhibitors
DE10043667A1 (en) 2000-09-05 2002-03-14 Merck Patent Gmbh 2-guanidino-4-aryl-quinazolines
US6414016B1 (en) 2000-09-05 2002-07-02 Sucampo, A.G. Anti-constipation composition
DE10046993A1 (en) 2000-09-22 2002-04-11 Aventis Pharma Gmbh Substituted cinnamic acid guanidides, process for their preparation, their use as a medicament and medicament containing them
AU2002213048A1 (en) 2000-10-05 2002-04-15 Smith Kline Beecham Corporation Phosphate transport inhibitors
WO2002040039A2 (en) 2000-11-20 2002-05-23 Dow Global Technologies Inc. In vivo use of water absorbent polymers
DE10063294A1 (en) 2000-12-19 2002-07-04 Aventis Pharma Gmbh New N-(heterocyclyl-alkyl)-fused norbornylamine derivatives, are NHE3 inhibitors useful e.g. for treating respiratory disorders, renal failure, intestinal dysfunction, lipid metabolism disorders or proliferative diseases
CN103638514A (en) 2001-03-29 2014-03-19 药物协和公司 Guanylate cyclase receptor agonists for the treatment of tissue inflammation and carcinogenesis
US6736705B2 (en) 2001-04-27 2004-05-18 Hitachi Global Storage Technologies Polishing process for glass or ceramic disks used in disk drive data storage devices
MY134211A (en) 2001-05-18 2007-11-30 Smithkline Beecham Corp Novel use
JP2007131532A (en) 2001-09-28 2007-05-31 Kirin Brewery Co Ltd Compound for inhibiting delivery of phosphorus in living body and drug containing the same
FR2830451B1 (en) 2001-10-09 2004-04-30 Inst Nat Sante Rech Med USE OF THYMULIN-LIKE PEPTIDES (PAT) FOR THE MANUFACTURE OF PAIN MEDICATIONS
JPWO2003048134A1 (en) 2001-12-05 2005-04-14 日本たばこ産業株式会社 Triazole compounds and pharmaceutical use thereof
US6911453B2 (en) 2001-12-05 2005-06-28 Aventis Pharma Deutschland Gmbh Substituted 4-phenyltetrahydroisoquinolinium, process for their preparation, their use as a medicament, and medicament containing them
OA12740A (en) * 2001-12-05 2006-06-30 Aventis Pharma Gmbh Substituted 4-phenyltetrahydroisoquinolines, method for the production thereof, the use thereof as medicaments, in addition to a medicament containingsame.
DE10161767A1 (en) 2001-12-15 2003-06-26 Merck Patent Gmbh New 2-guanidino-4-heterocyclyl-quinazoline derivatives, useful as sodium-proton antiporter subtype III inhibitors for treating e.g. respiratory, renal, ischemic or lipid metabolism disorders
EP1321142A1 (en) 2001-12-21 2003-06-25 Novartis AG Solid pharmaceutical composition for oral administration of Tegaserod
US6703405B2 (en) 2001-12-22 2004-03-09 Aventis Pharma Deutschland Gmbh Substituted 4-phenyltetrahydroisoquinolinium salts, process for their preparation, their use as a medicament, and medicament containing them
DE10163914A1 (en) * 2001-12-22 2003-07-03 Aventis Pharma Gmbh Substituted 4-phenyltetrahydroisoquinolinium salts, process for their preparation, their use as medicament, and medicament containing them
DE10163992A1 (en) 2001-12-24 2003-07-03 Merck Patent Gmbh 4-aryl-quinazolines
US7119120B2 (en) 2001-12-26 2006-10-10 Genzyme Corporation Phosphate transport inhibitors
ES2399880T3 (en) 2002-03-15 2013-04-04 Cypress Bioscience, Inc. Milnacipran for the treatment of irritable bowel syndrome
JP4465193B2 (en) 2002-03-19 2010-05-19 ジェンザイム コーポレーション Phosphate transport inhibitor
PL371596A1 (en) 2002-05-17 2005-06-27 Merck Patent Gmbh Use of compounds that are effective as selective opiate receptor modulators
US7014862B2 (en) 2002-05-20 2006-03-21 The Procter & Gamble Company Chewable compositions containing a gel-forming extract of psyllium
TWI245308B (en) 2002-09-13 2005-12-11 Getters Spa Non-evaporable getter compositions which can be reactivated at low temperature after exposure to reactive gases at a higher temperature
CN1254051C (en) 2002-10-25 2006-04-26 华为技术有限公司 User's data service sub speed delivering method in metropolitan area network transmission equipment
US6923466B2 (en) 2002-12-17 2005-08-02 James Tsai Collapsible handcart capable of extending the area of carrier by operating handle
US7304036B2 (en) 2003-01-28 2007-12-04 Microbia, Inc. Methods and compositions for the treatment of gastrointestinal disorders
US20050054705A1 (en) 2003-02-04 2005-03-10 Aventis Pharma Deutschland Gmbh N-substituted (benzoimidazol-2-yl) phenylamines, process for their preparation, their use as medicament or diagnostic aid, and medicament comprising them
US7026303B2 (en) 2003-02-18 2006-04-11 The Procter & Gamble Company Compositions comprising a polysaccharide component and one or more coating layers
WO2004085448A2 (en) 2003-03-19 2004-10-07 Genzyme Corporation Unsaturated phosphinyl-phosphonate phosphate transport inhibitors
DE10312963A1 (en) * 2003-03-24 2004-10-07 Aventis Pharma Deutschland Gmbh Substituted 4-phenyltetrahydroisoquinolines, process for their preparation, their use as medicament, and medicament containing them
US7241775B2 (en) 2003-03-24 2007-07-10 Sanofi-Aventis Deutschland Gmbh Composition, process of making, and medical use of substituted 4-phenyltetrahydroisoquinolines
US8134015B2 (en) 2003-03-27 2012-03-13 Kyowa Hakko Kirin Co., Ltd. Compound inhibiting in vivo phosphorous transport and medicine containing the same
DE112004001165T5 (en) 2003-06-25 2006-05-04 Cmet Inc., Yokohama An actinic radiation-curable stereolithographic resin composition having improved stability
WO2005020612A1 (en) 2003-08-20 2005-03-03 Musky Communications (Proprietary) Limited Telephonic communication
PE20050952A1 (en) * 2003-09-24 2005-12-19 Novartis Ag DERIVATIVES OF ISOQUINOLINE AS INHIBITORS OF B-RAF
US20050095578A1 (en) 2003-10-31 2005-05-05 Koller Manfred R. Method and apparatus for cell permeabilization
WO2005054705A2 (en) 2003-11-26 2005-06-16 Brian Judd Apparatus for dampening impact vibration in manual tools
US20050244367A1 (en) 2004-05-03 2005-11-03 Ilypsa, Inc. Phospholipase inhibitors localized in the gastrointestinal lumen
CA2567470A1 (en) 2004-05-14 2006-01-05 The University Of North Carolina At Chapel Hill Prouroguanylin as therapeutic and diagnostic agents
US7598863B2 (en) 2004-05-14 2009-10-06 Paceco Corp. Method and apparatus for making status reporting devices for container handlers
DE102004046492A1 (en) * 2004-09-23 2006-03-30 Sanofi-Aventis Deutschland Gmbh Substituted 4-phenyltetrahydroisoquinolines, process for their preparation, their use as medicament, and medicament containing them
US20070054916A1 (en) * 2004-10-01 2007-03-08 Amgen Inc. Aryl nitrogen-containing bicyclic compounds and methods of use
EP1799791B1 (en) 2004-10-13 2016-01-13 Rolic AG Photocrosslinkable materials
DE102004054847A1 (en) 2004-11-13 2006-05-24 Sanofi-Aventis Deutschland Gmbh Substituted benzoylguanidines, process for their preparation, their use as medicament or diagnostic agent, and medicament containing them
DE102005001411A1 (en) * 2005-01-12 2006-07-27 Sanofi-Aventis Deutschland Gmbh Substituted 4-phenyltetrahydroisoquinolines, process for their preparation, their use as medicament, and medicament containing them
CA2619977C (en) 2005-08-29 2011-06-28 Demain Technology Pty Ltd. Power tool
DE102005044815A1 (en) 2005-09-20 2007-03-22 Sanofi-Aventis Deutschland Gmbh Use of inhibitors of Na + / H + exchanger, subtype 5 (NHE5) for memory improvement
DE102005044817A1 (en) * 2005-09-20 2007-03-22 Sanofi-Aventis Deutschland Gmbh Substituted 4-phenyltetrahydroisoquinolines, process for their preparation, their use as medicament, and medicament containing them
WO2007056281A2 (en) 2005-11-03 2007-05-18 Ilypsa, Inc. Multivalent indole compounds and use thereof as phospholipase-a2 inhibitors
WO2007056279A2 (en) * 2005-11-03 2007-05-18 Ilypsa, Inc. Phospholipase inhibitors, including multi-valent phospholipase inhibitors, and use thereof, including as lumen-localized phospholipase inhibitors
US7694181B2 (en) 2005-12-12 2010-04-06 Archivas, Inc. Automated software testing framework
GB0610009D0 (en) 2006-05-19 2006-06-28 Health Prot Agency Culture medium
GB2438389A (en) 2006-05-23 2007-11-28 Gkn Aerospace Transparency Sys Heating system for leading edge of aircraft
US20120283411A9 (en) 2006-06-29 2012-11-08 Ironwood Pharmaceuticals, Inc. Methods and compositions for the treatment of gastrointestinal disorders
US8617903B2 (en) 2007-01-29 2013-12-31 The Invention Science Fund I, Llc Methods for allergen detection
EP2129683A4 (en) 2007-02-26 2011-01-05 Ironwood Pharmaceuticals Inc Methods and compositions for the treatment of heart failure and other disorders
EP2671584A3 (en) 2007-05-04 2014-03-26 Ironwood Pharmaceuticals, Inc. Compositions and methods for treating disorders associated with salt or fluid retention
KR20170091783A (en) 2008-12-31 2017-08-09 알데릭스, 인코포레이티드 Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11318129B2 (en) 2008-12-31 2022-05-03 Ardelyx, Inc. Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
US10543207B2 (en) 2008-12-31 2020-01-28 Ardelyx, Inc. Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
CN102757390A (en) * 2011-04-27 2012-10-31 中国中化股份有限公司 Method for preparing 2-methoxy-4-diazanyl-5-fluoropyrimidine
CN104902930A (en) * 2012-08-21 2015-09-09 阿德利克斯公司 Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
US20170368004A1 (en) * 2012-08-21 2017-12-28 Ardelyx, Inc Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
US10385024B2 (en) 2012-08-21 2019-08-20 Ardelyx, Inc. Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
US20150299131A1 (en) * 2012-08-21 2015-10-22 Ardelyx, Inc Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
US20150336892A1 (en) * 2012-08-21 2015-11-26 Ardelyx, Inc Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
AU2013304812B2 (en) * 2012-08-21 2016-06-09 Ardelyx, Inc. Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
AU2013304813B2 (en) * 2012-08-21 2016-08-04 Ardelyx, Inc. Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
KR20150073162A (en) * 2012-08-21 2015-06-30 알데릭스, 인코포레이티드 Compounds and methods for inhibiting nhe-mediaded antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
WO2014029983A1 (en) 2012-08-21 2014-02-27 Ardelyx, Inc. Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
KR20150073949A (en) * 2012-08-21 2015-07-01 알데릭스, 인코포레이티드 Compounds and methods for inhibiting nhe-mediaded antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
US10376481B2 (en) * 2012-08-21 2019-08-13 Ardelyx, Inc. Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
RU2684097C2 (en) * 2012-08-21 2019-04-04 Эрделикс, Инк. Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal diseases
KR102138390B1 (en) * 2012-08-21 2020-07-27 알데릭스, 인코포레이티드 Compounds and methods for inhibiting nhe-mediaded antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
KR102138391B1 (en) * 2012-08-21 2020-07-27 알데릭스, 인코포레이티드 Compounds and methods for inhibiting nhe-mediaded antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
WO2014029984A1 (en) * 2012-08-21 2014-02-27 Ardelyx, Inc. Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
US10940146B2 (en) 2013-04-12 2021-03-09 Ardelyx, Inc. NHE3-binding compounds and methods for inhibiting phosphate transport
US10272079B2 (en) 2013-04-12 2019-04-30 Ardelyx, Inc. NHE3-binding compounds and methods for inhibiting phosphate transport
EP3552630A1 (en) * 2013-04-12 2019-10-16 Ardelyx, Inc. Nhe3-binding compounds for inhibiting phosphate transport
EP3988120A1 (en) * 2013-04-12 2022-04-27 Ardelyx, Inc. Nhe3-binding compounds and methods for inhibiting phosphate transport
EP3173408A4 (en) * 2014-07-25 2017-12-06 Taisho Pharmaceutical Co., Ltd. Phenyl tetrahydroisoquinoline compound substituted with heteroaryl
KR20170034897A (en) 2014-07-25 2017-03-29 다이쇼 세이야꾸 가부시끼가이샤 Phenyl tetrahydroisoquinoline compound substituted with heteroaryl
US9932331B2 (en) 2014-07-25 2018-04-03 Taisho Pharmaceutical Co., Ltd. Phenyl tetrahydroisoquinoline compound substituted with heteroaryl
WO2018129552A1 (en) 2017-01-09 2018-07-12 Ardelyx, Inc. Compounds useful for treating gastrointestinal tract disorders
WO2018129556A1 (en) * 2017-01-09 2018-07-12 Ardelyx, Inc. Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
WO2018129557A1 (en) 2017-01-09 2018-07-12 Ardelyx, Inc. Inhibitors of nhe-mediated antiport
IL267821B2 (en) * 2017-01-09 2023-02-01 Ardelyx Inc Inhibitors of nhe-mediated antiport
IL267821B (en) * 2017-01-09 2022-10-01 Ardelyx Inc Inhibitors of nhe-mediated antiport
AU2018206479B2 (en) * 2017-01-09 2022-07-14 Ardelyx, Inc. Inhibitors of NHE-mediated antiport
US11147884B2 (en) 2017-01-09 2021-10-19 Ardelyx, Inc. Inhibitors of NHE-mediated antiport
US11242337B2 (en) 2017-01-09 2022-02-08 Ardelyx, Inc. Compounds useful for treating gastrointestinal tract disorders
US11938134B2 (en) 2017-03-10 2024-03-26 Eikonizo Therapeutics, Inc. Metalloenzyme inhibitor compounds
WO2019060051A1 (en) 2017-08-04 2019-03-28 Ardelyx, Inc. Glycyrrhetinic acid derivatives for treating hyperkalemia
WO2019091503A1 (en) 2017-11-13 2019-05-16 Zentiva K.S. Solid forms of tenapanor and method of preparation of tenapanor
WO2020051014A1 (en) 2018-09-04 2020-03-12 Teva Pharmaceuticals International Gmbh Processes for the preparation of tenapanor and intermediates thereof
WO2020163642A1 (en) 2019-02-07 2020-08-13 Ardelyx, Inc. Glycyrrhetinic acid derivatives for use in treating hyperkalemia
EP4234016A2 (en) 2019-02-07 2023-08-30 Ardelyx, Inc. Glycyrrhetinic acid derivatives for use in treating hyperkalemia
WO2020231770A1 (en) 2019-05-16 2020-11-19 Eli Lilly And Company Sodium-hydrogen exchanger 3 inhibitor compounds
WO2020237096A1 (en) 2019-05-21 2020-11-26 Ardelyx, Inc. Combination for lowering serum phosphate in a patient
CN112679427A (en) * 2019-10-17 2021-04-20 江苏恒瑞医药股份有限公司 Tetrahydroisoquinoline derivatives, preparation method and medical application thereof
CN112047881A (en) * 2020-10-10 2020-12-08 山东汇海医药化工有限公司 Preparation method of tanaprost
CN112047881B (en) * 2020-10-10 2023-04-11 山东汇海医药化工有限公司 Preparation method of tanaprost

Also Published As

Publication number Publication date
AU2009334511A1 (en) 2011-07-21
KR20170091783A (en) 2017-08-09
AU2009334511C1 (en) 2016-08-18
CN103819403A (en) 2014-05-28
WO2010078449A3 (en) 2010-09-30
MX345283B (en) 2017-01-24
CA2748607A1 (en) 2010-07-08
US20150190389A1 (en) 2015-07-09
BRPI0923861B8 (en) 2021-05-25
KR20220042487A (en) 2022-04-05
US20140107074A1 (en) 2014-04-17
BRPI0923861B1 (en) 2020-03-10
IL250641A0 (en) 2017-04-30
PL2384318T3 (en) 2018-04-30
KR20110110287A (en) 2011-10-06
EP3351248B1 (en) 2021-06-09
PT2384318T (en) 2018-02-07
IL250641B (en) 2018-06-28
IL213852A0 (en) 2011-07-31
US9006281B2 (en) 2015-04-14
IL259851A (en) 2018-07-31
HK1198162A1 (en) 2015-03-13
IL259851B (en) 2020-07-30
JP2014114300A (en) 2014-06-26
MX2011007024A (en) 2011-09-27
KR101766619B1 (en) 2017-08-08
EP3939964A1 (en) 2022-01-19
SI2384318T1 (en) 2018-03-30
CY1120451T1 (en) 2019-07-10
US20130274285A1 (en) 2013-10-17
ES2657938T3 (en) 2018-03-07
HUE036405T2 (en) 2018-07-30
IL213852A (en) 2017-02-28
HRP20180289T1 (en) 2018-03-23
JP5502106B2 (en) 2014-05-28
KR101683318B1 (en) 2016-12-07
JP5805802B2 (en) 2015-11-10
KR20160140994A (en) 2016-12-07
EP3351248A1 (en) 2018-07-25
US8969377B2 (en) 2015-03-03
EP2384318A2 (en) 2011-11-09
US8541448B2 (en) 2013-09-24
LT2384318T (en) 2018-02-26
CN103819403B (en) 2017-01-04
US20120263670A1 (en) 2012-10-18
CN102333759A (en) 2012-01-25
KR20200111230A (en) 2020-09-28
EP2384318B1 (en) 2017-11-15
DK2384318T3 (en) 2018-02-19
NO2384318T3 (en) 2018-04-14
BRPI0923861A2 (en) 2015-07-28
JP2012514009A (en) 2012-06-21
US9408840B2 (en) 2016-08-09
AU2009334511B2 (en) 2016-01-28

Similar Documents

Publication Publication Date Title
AU2009334511C1 (en) Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
US11318129B2 (en) Compounds and methods for inhibiting NHE-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
EP2887963B1 (en) Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
EP2887964B1 (en) Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders
WO2018129556A1 (en) Compounds and methods for inhibiting nhe-mediated antiport in the treatment of disorders associated with fluid retention or salt overload and gastrointestinal tract disorders

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200980157614.8

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09796924

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2009334511

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2011543730

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2748607

Country of ref document: CA

Ref document number: MX/A/2011/007024

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 5002/CHENP/2011

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2009334511

Country of ref document: AU

Date of ref document: 20091230

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20117017941

Country of ref document: KR

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2009796924

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2009796924

Country of ref document: EP

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: PI0923861

Country of ref document: BR

ENP Entry into the national phase

Ref document number: PI0923861

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20110628

WWE Wipo information: entry into national phase

Ref document number: 250641

Country of ref document: IL